101
|
Entropy as a Novel Measure of Myocardial Tissue Heterogeneity for Prediction of Ventricular Arrhythmias and Mortality in Post-Infarct Patients. JACC Clin Electrophysiol 2019; 5:480-489. [DOI: 10.1016/j.jacep.2018.12.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 12/12/2018] [Accepted: 12/12/2018] [Indexed: 11/24/2022]
|
102
|
Bax JJ, Di Carli M, Narula J, Delgado V. Multimodality imaging in ischaemic heart failure. Lancet 2019; 393:1056-1070. [PMID: 30860031 DOI: 10.1016/s0140-6736(18)33207-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 12/01/2018] [Accepted: 12/07/2018] [Indexed: 12/30/2022]
Abstract
In heart failure, extensive evaluation with modern non-invasive imaging modalities is needed to assess causes, pathophysiology, and haemodynamics, to determine prognosis and consider therapeutic options. This systematic evaluation includes a stepwise assessment of left ventricular size and function, the presence and severity of coronary artery disease, mitral regurgitation, pulmonary hypertension, right ventricular dilation and dysfunction, and tricuspid regurgitation. Based on this imaging-derived information, the need for specific therapies besides optimised medical therapy can be determined. The need for revascularisation, implantation of an implantable cardiac defibrillator, and mitral or tricuspid valve repair or replacement, can be (partially) guided by non-invasive imaging. Importantly, randomised controlled trials on the use of non-inasive imaging to guide therapy are scarce in this field and most non-pharmacological therapies are based on expert-consensus, but whenever trials are available, they will be addressed in this paper.
Collapse
Affiliation(s)
- Jeroen J Bax
- Department of Cardiology, Leiden University Medical Center, Leiden, Netherlands.
| | - Marcelo Di Carli
- Departments of Radiology and Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, NY, USA
| | - Jagat Narula
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Victoria Delgado
- Department of Cardiology, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
103
|
Storz C, Hetterich H, Lorbeer R, Heber SD, Schafnitzel A, Patscheider H, Auweter S, Zitzelsberger T, Rathmann W, Nikolaou K, Reiser M, Schlett CL, von Knobelsdorff-Brenkenhoff F, Peters A, Schulz-Menger J, Bamberg F. Myocardial tissue characterization by contrast-enhanced cardiac magnetic resonance imaging in subjects with prediabetes, diabetes, and normal controls with preserved ejection fraction from the general population. Eur Heart J Cardiovasc Imaging 2019; 19:701-708. [PMID: 28950340 DOI: 10.1093/ehjci/jex190] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 07/10/2017] [Indexed: 01/18/2023] Open
Abstract
Aims To characterize changes in the myocardium in subjects with prediabetes, diabetes, and healthy controls with preserved left ventricular ejection fraction (LVEF) by using cardiac magnetic resonance imaging (CMR) in a sample from the general population. Methods and results Subjects without history of cardiovascular disease and preserved LVEF but established diabetes, prediabetes, and controls from a population-based cohort underwent contrast-enhanced CMR. Obtained parameters included left ventricular (LV) function and morphology, late gadolinium enhancement as well as T1-mapping and derivation of extracellular volume fraction (ECV) by modified Look-Locker inversion recovery for diffuse fibrosis in a subset of patients. Fibrosis volume and cell volume were calculated and LV remodelling index was calculated by dividing the LV mass by its end-diastolic volume. Among 343 subjects (56.1 ± 9.2 years, 57% males), 47 subjects were classified as diabetes, 78 as prediabetes, and 218 as controls. Haematocrit values and thus ECV parameters were available in 251 subjects. LV remodelling index was significantly higher in participants with prediabetes and diabetes, independent of body mass index (BMI), hypertension, age, and sex. ECV was decreased in subjects with prediabetes and diabetes compared with healthy controls (23.1 ± 2.4% and 22.8 ± 3.0%, both P < 0.007). In contrast, cell volume was significantly higher in subjects with prediabetes and diabetes as compared with controls (109.1 ± 23.8 and 114.9 ± 32.3 mL vs. 96.5 ± 26.9 mL, both P < 0.03, respectively). However, differences in ECV and cell volume attenuated after the adjustment for cardiometabolic risk factors, including age, sex, BMI, and hypertension. Conclusion Subjects with prediabetes and diabetes but preserved LVEF had higher LV remodelling indices, suggesting early detectable changes in the disease process, while diffuse myocardial fibrosis appears to be less relevant at this stage.
Collapse
Affiliation(s)
- Corinna Storz
- Department of Diagnostic and Interventional Radiology, University of Tuebingen, Hoppe-Seyler-Straße 3, 72076 Tuebingen, Germany
| | - Holger Hetterich
- Institute of Clinical Radiology, Ludwig-Maximilians-University Hospital, Marchioninistraße 15, Munich 81377, Germany
| | - Roberto Lorbeer
- Institute of Clinical Radiology, Ludwig-Maximilians-University Hospital, Marchioninistraße 15, Munich 81377, Germany
| | - Sophia D Heber
- Department of Diagnostic and Interventional Radiology, University of Tuebingen, Hoppe-Seyler-Straße 3, 72076 Tuebingen, Germany
| | - Anina Schafnitzel
- Institute of Clinical Radiology, Ludwig-Maximilians-University Hospital, Marchioninistraße 15, Munich 81377, Germany
| | - Hanna Patscheider
- Institute of Clinical Radiology, Ludwig-Maximilians-University Hospital, Marchioninistraße 15, Munich 81377, Germany
| | - Sigrid Auweter
- Institute of Clinical Radiology, Ludwig-Maximilians-University Hospital, Marchioninistraße 15, Munich 81377, Germany
| | - Tanja Zitzelsberger
- Department of Diagnostic and Interventional Radiology, University of Tuebingen, Hoppe-Seyler-Straße 3, 72076 Tuebingen, Germany
| | - Wolfgang Rathmann
- Department of Biometry and Epidemiology, German Diabetes Center, Auf'm Hennekamp 65, Duesseldorf 40225, Germany
| | - Konstantin Nikolaou
- Department of Diagnostic and Interventional Radiology, University of Tuebingen, Hoppe-Seyler-Straße 3, 72076 Tuebingen, Germany
| | - Maximilian Reiser
- Institute of Clinical Radiology, Ludwig-Maximilians-University Hospital, Marchioninistraße 15, Munich 81377, Germany
| | - Christopher L Schlett
- Department of Diagnostic and Interventional Radiology, University Hospital Heidelberg, Im Neuenheimer Feld 400, Heidelberg 69120, Germany
| | - Florian von Knobelsdorff-Brenkenhoff
- Department of Cardiology, Charité, Experimental and Clinical Research Center and HELIOS-Clinics Berlin-Buch Schwanebecker Chaussee 50, 13125 Berlin, Germany.,Department of Cardiology, Clinic Agatharied, Ludwig-Maximilians-University Munich, Norbert-Kerkel-Platz, Hausham 83734, Germany
| | - Annette Peters
- Institute of Epidemiology II, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstraße 1, 85764 Neuherberg, Germany.,Institute for Cardiovascular Prevention, Ludwig-Maximilian-University-Hospital, Pettenkoferstraäe 9, Munich 80336, Germany.,German Center for Cardiovascular Disease Research (DZHK e.V.), Partnersite Munich, Biedersteiner Straße 29, Munich 80802, Germany
| | - Jeanette Schulz-Menger
- Department of Cardiology, Charité, Experimental and Clinical Research Center and HELIOS-Clinics Berlin-Buch Schwanebecker Chaussee 50, 13125 Berlin, Germany.,German Center for Cardiovascular Disease Research (DZHK e.V.), Partnersite Berlin, Oudenarder Straße 16, Berlin 13347, Germany
| | - Fabian Bamberg
- Department of Diagnostic and Interventional Radiology, University of Tuebingen, Hoppe-Seyler-Straße 3, 72076 Tuebingen, Germany
| |
Collapse
|
104
|
Incremental value of extracellular volume assessment by cardiovascular magnetic resonance imaging in risk stratifying patients with suspected myocarditis. Int J Cardiovasc Imaging 2019; 35:1067-1078. [DOI: 10.1007/s10554-019-01552-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 02/02/2019] [Indexed: 01/27/2023]
|
105
|
Shanbhag SM, Greve AM, Aspelund T, Schelbert EB, Cao JJ, Danielsen R, þorgeirsson G, Sigurðsson S, Eiríksdóttir G, Harris TB, Launer LJ, Guðnason V, Arai AE. Prevalence and prognosis of ischaemic and non-ischaemic myocardial fibrosis in older adults. Eur Heart J 2019; 40:529-538. [PMID: 30445559 PMCID: PMC6657269 DOI: 10.1093/eurheartj/ehy713] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 10/25/2017] [Accepted: 10/11/2018] [Indexed: 12/23/2022] Open
Abstract
Aims Non-ischaemic cardiomyopathies (NICM) can cause heart failure and death. Cardiac magnetic resonance (CMR) detects myocardial scar/fibrosis associated with myocardial infarction (MI) and NICM with late gadolinium enhancement (LGE). The aim of this study was to determine the prevalence and prognosis of ischaemic and non-ischaemic myocardial fibrosis in a community-based sample of older adults. Methods and results The ICELAND-MI cohort, a substudy of the Age, Gene/Environment Susceptibility Reykjavik (AGES-Reykjavik) study, provided a well-characterized population of 900 subjects after excluding subjects with pre-existing heart failure. Late gadolinium enhancement CMR divided subjects into four groups: MI (n = 211), major (n = 54) non-ischaemic fibrosis (well-established, classic patterns, associated with myocarditis, infiltrative cardiomyopathies, or pathological hypertrophy), minor (n = 238) non-ischaemic fibrosis (remaining localized patterns not meeting major criteria), and a no LGE (n = 397) reference group. The primary outcome was time to death or first heart failure hospitalization. During a median follow-up of 5.8 years, 192 composite events occurred (115 deaths and 77 hospitalizations for incident heart failure). After inverse probability weighting, major non-ischaemic fibrosis [hazard ratio (HR) 3.2, P < 0.001] remained independently associated with the primary endpoint, while MI (HR 1.4, P = 0.10) and minor non-ischaemic LGE (HR 1.2, P = 0.39) did not. Major non-ischaemic fibrosis was associated with a poorer outcome than MI (HR = 2.3, P = 0.001) in the adjusted analysis. Conclusion Major non-ischaemic patterns of myocardial fibrosis portended worse prognosis than no fibrosis/scar in an older community-based cohort. Traditional risk factors largely accounted for the effect of MI and minor non-ischaemic LGE.
Collapse
Affiliation(s)
- Sujata M Shanbhag
- Department of Health and Human Services, National Heart, Lung, and Blood Institute, National Institutes of Health, Building 10, Room B1D416, MSC 1061, 10 Center Dr., Bethesda, MD, USA
| | - Anders M Greve
- Department of Health and Human Services, National Heart, Lung, and Blood Institute, National Institutes of Health, Building 10, Room B1D416, MSC 1061, 10 Center Dr., Bethesda, MD, USA
- Department of Clinical Biochemistry, Rigshospitalet, 9 Blegdamsvej, Copenhagen, Denmark
| | - Thor Aspelund
- Hjartavernd (Icelandic Heart Association), Holtasmari 1, Kopavogur, Iceland
- University of Iceland, Sæmundargata 2, Reykjavik, Iceland
| | - Erik B Schelbert
- Department of Health and Human Services, National Heart, Lung, and Blood Institute, National Institutes of Health, Building 10, Room B1D416, MSC 1061, 10 Center Dr., Bethesda, MD, USA
- University of Pittsburgh Medical Center, Heart and Vascular Institute, 200 Lothrop St., Ste. A349, Pittsburgh, PA, USA
| | - J Jane Cao
- Department of Health and Human Services, National Heart, Lung, and Blood Institute, National Institutes of Health, Building 10, Room B1D416, MSC 1061, 10 Center Dr., Bethesda, MD, USA
- St. Francis Hospital, The heart Center, State University of New York at Stony Brook, 100 Port Washington Blvd, Roslyn, NY, USA
| | | | | | | | - Guðný Eiríksdóttir
- Hjartavernd (Icelandic Heart Association), Holtasmari 1, Kopavogur, Iceland
| | - Tamara B Harris
- Laboratory of Epidemiology & Population Science, National Institute on Aging, National Institutes of Health, Department of Health and Human Services, GWY Bldg Rm 2N300, 7201 Wisconsin Ave, Bethesda, MD, USA
| | - Lenore J Launer
- Laboratory of Epidemiology & Population Science, National Institute on Aging, National Institutes of Health, Department of Health and Human Services, GWY Bldg Rm 2N300, 7201 Wisconsin Ave, Bethesda, MD, USA
| | - Vilmundur Guðnason
- Hjartavernd (Icelandic Heart Association), Holtasmari 1, Kopavogur, Iceland
- University of Iceland, Sæmundargata 2, Reykjavik, Iceland
| | - Andrew E Arai
- Department of Health and Human Services, National Heart, Lung, and Blood Institute, National Institutes of Health, Building 10, Room B1D416, MSC 1061, 10 Center Dr., Bethesda, MD, USA
| |
Collapse
|
106
|
Affiliation(s)
- A Mark Richards
- From the Cardiovascular Research Institute, National University Heart Centre, Singapore; and Christchurch Heart Institute, University of Otago, Dunedin, New Zealand.
| |
Collapse
|
107
|
Extracellular volume by cardiac magnetic resonance is associated with biomarkers of inflammation in hypertensive heart disease. J Hypertens 2019; 37:65-72. [DOI: 10.1097/hjh.0000000000001875] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
108
|
Ferreira VM, Piechnik SK. T1 or ECV?: Depends on the Methods. JACC Cardiovasc Imaging 2018; 12:1670-1672. [PMID: 30448140 DOI: 10.1016/j.jcmg.2018.10.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 10/02/2018] [Indexed: 12/23/2022]
Affiliation(s)
- Vanessa M Ferreira
- Oxford Centre for Clinical Magnetic Resonance Research, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom.
| | - Stefan K Piechnik
- Oxford Centre for Clinical Magnetic Resonance Research, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
109
|
Brown PF, Miller C, Di Marco A, Schmitt M. Towards cardiac MRI based risk stratification in idiopathic dilated cardiomyopathy. Heart 2018; 105:270-275. [PMID: 30377260 DOI: 10.1136/heartjnl-2018-313767] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2018] [Revised: 10/02/2018] [Accepted: 10/04/2018] [Indexed: 12/29/2022] Open
Abstract
Sudden cardiac death (SCD) secondary to arrhythmia remains a risk in those with dilated cardiomyopathy (DCM), an implantable cardiac defibrillator (ICD) is an effective strategy to prevent SCD. Current guidelines recommend selection for ICD based on ejection fraction (EF) less than 35%, however, most SCD occurs in those with EF>35%. Although meta-analysis has demonstrated a survival benefit for primary prevention ICD in DCM, no randomised trial has shown a significant reduction in overall mortality including the most recent 'Danish Study to Assess the Efficacy of ICDs in Patients With Non-Ischemic Systolic Heat Failure on Mortality' study. Clearly, a more sophisticated selection strategy is required. Cardiac MRI (CMR) is an ideal non-invasive imaging technique which allows calculation of EF as well as tissue characterisation with gadolinium contrast, parametric mapping and feature tracking. Late gadolinium enhancement detects mid-wall fibrosis in approximately 30% of those with DCM, three meta-analyses have demonstrated an association between fibrosis in DCM and SCD, and those without fibrosis are at low risk of SCD. T1 mapping and extracellular volume (ECV) calculation are methods of demonstrating diffuse fibrosis in the myocardium. Raised ECV and native T1 have been associated with worse outcomes but the relationship to SCD has not been well studied. Undoubtedly, more research is required but CMR has several tools which offer incremental value above EF to improve risk stratification and consequent outcomes and resource utilisation in those with DCM.
Collapse
Affiliation(s)
- Pamela Frances Brown
- Cardiac MRI Department, North West Heart Centre, Manchester University Foundation Trust-Wythenshawe Site, Manchester, UK
| | - Chris Miller
- Cardiac MRI Department, North West Heart Centre, Manchester University Foundation Trust-Wythenshawe Site, Manchester, UK
| | - Andrea Di Marco
- Arrhythmia Unit, Heart Disease Institute, Bellvitge University Hospital, Barcelona, Spain
| | - Matthias Schmitt
- Cardiac MRI Department, North West Heart Centre, Manchester University Foundation Trust-Wythenshawe Site, Manchester, UK
| |
Collapse
|
110
|
Gorcsan J, Kagiyama N. Can global longitudinal strain predict response to cardiac resynchronization therapy? Heart Rhythm 2018; 15:1540-1541. [DOI: 10.1016/j.hrthm.2018.05.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Indexed: 10/14/2022]
|
111
|
Habib G, Bucciarelli-Ducci C, Caforio ALP, Cardim N, Charron P, Cosyns B, Dehaene A, Derumeaux G, Donal E, Dweck MR, Edvardsen T, Erba PA, Ernande L, Gaemperli O, Galderisi M, Grapsa J, Jacquier A, Klingel K, Lancellotti P, Neglia D, Pepe A, Perrone-Filardi P, Petersen SE, Plein S, Popescu BA, Reant P, Sade LE, Salaun E, Slart RHJA, Tribouilloy C, Zamorano J. Multimodality Imaging in Restrictive Cardiomyopathies: An EACVI expert consensus document In collaboration with the "Working Group on myocardial and pericardial diseases" of the European Society of Cardiology Endorsed by The Indian Academy of Echocardiography. Eur Heart J Cardiovasc Imaging 2018; 18:1090-1121. [PMID: 28510718 DOI: 10.1093/ehjci/jex034] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 02/14/2017] [Indexed: 12/11/2022] Open
Abstract
Restrictive cardiomyopathies (RCMs) are a diverse group of myocardial diseases with a wide range of aetiologies, including familial, genetic and acquired diseases and ranging from very rare to relatively frequent cardiac disorders. In all these diseases, imaging techniques play a central role. Advanced imaging techniques provide important novel data on the diagnostic and prognostic assessment of RCMs. This EACVI consensus document provides comprehensive information for the appropriateness of all non-invasive imaging techniques for the diagnosis, prognostic evaluation, and management of patients with RCM.
Collapse
Affiliation(s)
- Gilbert Habib
- Aix- Aix-Marseille Univ, URMITE, Aix Marseille Université-UM63, CNRS 7278, IRD 198, INSERM 1095.,Cardiology Department, APHM, La Timone Hospital, Boulevard Jean Moulin, 13005 Marseille, France
| | - Chiara Bucciarelli-Ducci
- Bristol Heart Institute, National Institute of Health Research (NIHR) Bristol Cardiovascular Biomedical Research Unit (BRU), University of Bristol, Bristol, UK
| | - Alida L P Caforio
- Cardiology, Department of Cardiological Thoracic and Vascular Sciences, University of Padova, Italy
| | - Nuno Cardim
- Multimodality Cardiac Imaging Department, Sports Cardiology and Cardiomyopathies Centre-Hospital da Luz; Lisbon, Portugal
| | - Philippe Charron
- Université Versailles Saint Quentin, INSERM U1018, Hôpital Ambroise Paré, Boulogne-Billancourt, France.,Centre de référence pour les maladies cardiaques héréditaires, APHP, ICAN, Hôpital de la Pitié Salpêtrière, Paris, France
| | | | - Aurélie Dehaene
- Department of Radiology and Cardiovascular Imaging, APHM, Hôpitaux de la Timone, Pôle d'imagerie Médicale, 13005 Marseille, France
| | - Genevieve Derumeaux
- Department of Physiology, INSERM U955, Université Paris-Est Creteil, Henri Mondor Hospital, DHU-ATVB, AP-HP, Créteil, France
| | - Erwan Donal
- Cardiologie-CHU Rennes & CIC-IT 1414 & LTSI INSERM 1099 - Université Rennes-1
| | - Marc R Dweck
- Centre for Cardiovascular Science, University of Edinburgh
| | - Thor Edvardsen
- Department of Cardiology, Center for Cardiological Innovation and Institute for Surgical Research, Oslo University Hospital, Oslo, Norway.,University of Oslo, Oslo, Norway
| | - Paola Anna Erba
- Regional Center of Nuclear Medicine, Department of Translational Research and New Technology in Medicine, University of Pisa, Pisa, Italy
| | - Laura Ernande
- Department of Physiology, INSERM U955, Université Paris-Est Creteil, Henri Mondor Hospital, DHU-ATVB, AP-HP, Créteil, France
| | - Oliver Gaemperli
- University Heart Center Zurich, Interventional Cardiology and Cardiac Imaging 19, Zurich
| | - Maurizio Galderisi
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | - Julia Grapsa
- Department of Cardiovascular Sciences, Imperial College of London, London, UK
| | - Alexis Jacquier
- Department of Radiology and Cardiovascular Imaging, APHM, Hôpitaux de la Timone, Pôle d'imagerie Médicale, Aix-Marseille Université, CNRS, CRMBM UMR 7339, 13385 Marseille, France
| | - Karin Klingel
- Department of Molecular Pathology, Institute for Pathology and Neuropathology, University Hospital Tuebingen, Tuebingen, Germany
| | - Patrizio Lancellotti
- Departments of Cardiology, Heart Valve Clinic, University of Liège Hospital, GIGA Cardiovascular Sciences, CHU Sart Tilman, Liège, Belgium.,Gruppo Villa Maria Care and Research, Anthea Hospital, Bari, Italy
| | - Danilo Neglia
- Cardiovascular Department, Fondazione Toscana G. Monasterio, CNR Institute of Clinical Physiology, Scuola Superiore Sant'Anna, Pisa, Italy
| | - Alessia Pepe
- Magnetic Resonance Imaging Unit, Fondazione G. Monasterio C.N.R.-Regione Toscana Pisa, Italy
| | | | - Steffen E Petersen
- Department of Advanced Cardiovascular Imaging, William Harvey Research Institute, National Institute for Health Research Cardiovascular Biomedical Research Unit at Barts, London, UK
| | - Sven Plein
- Division of Biomedical Imaging, Multidisciplinary Cardiovascular Research Centre, Leeds Institute of Cardiovascular and Metabolic Medicine LIGHT Laboratories, University of Leeds, UK
| | - Bogdan A Popescu
- University of Medicine and Pharmacy 'Carol Davila'-Euroecolab, Institute of Cardiovascular Diseases, Bucharest, Romania
| | | | | | - Erwan Salaun
- Cardiology Department, La Timone Hospital, Marseille France
| | - Riemer H J A Slart
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Hanzeplein 1, Groningen, The Netherlands.,Department of Biomedical Photonic Imaging, University of Twente, PO Box 217, 7500 AEEnschede, The Netherlands
| | - Christophe Tribouilloy
- Department of Cardiology, University Hospital Amiens, Amiens, France and INSERM U-1088, Jules Verne University of Picardie, Amiens, France
| | - Jose Zamorano
- University Hospital Ramon y Cajal Carretera de Colmenar Km 9,100, 28034 Madrid, Spain
| | | | | |
Collapse
|
112
|
Prognostic value of T1 mapping and extracellular volume fraction in cardiovascular disease: a systematic review and meta-analysis. Heart Fail Rev 2018; 23:723-731. [DOI: 10.1007/s10741-018-9718-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
113
|
Engblom H, Kanski M, Kopic S, Nordlund D, Xanthis CG, Jablonowski R, Heiberg E, Aletras AH, Carlsson M, Arheden H. Importance of standardizing timing of hematocrit measurement when using cardiovascular magnetic resonance to calculate myocardial extracellular volume (ECV) based on pre- and post-contrast T1 mapping. J Cardiovasc Magn Reson 2018; 20:46. [PMID: 29950178 PMCID: PMC6022290 DOI: 10.1186/s12968-018-0464-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 05/24/2018] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND Cardiovascular magnetic resonance (CMR) can be used to calculate myocardial extracellular volume fraction (ECV) by relating the longitudinal relaxation rate in blood and myocardium before and after contrast-injection to hematocrit (Hct) in blood. Hematocrit is known to vary with body posture, which could affect the calculations of ECV. The aim of this study was to test the hypothesis that there is a significant increase in calculated ECV values if the Hct is sampled after the CMR examination in supine position compared to when the patient arrives at the MR department. METHODS Forty-three consecutive patients including various pathologies as well as normal findings were included in the study. Venous blood samples were drawn upon arrival to the MR department and directly after the examination with the patient remaining in supine position. A Modified Look-Locker Inversion recovery (MOLLI) protocol was used to acquire mid-ventricular short-axis images before and after contrast injection from which motion-corrected T1 maps were derived and ECV was calculated. RESULTS Hematocrit decreased from 44.0 ± 3.7% before to 40.6 ± 4.0% after the CMR examination (p < 0.001). This resulted in a change in calculated ECV from 24.7 ± 3.8% before to 26.2 ± 4.2% after the CMR examination (p < 0.001). All patients decreased in Hct after the CMR examination compared to before except for two patients whose Hct remained the same. CONCLUSION Variability in CMR-derived myocardial ECV can be reduced by standardizing the timing of Hct measurement relative to the CMR examination. Thus, a standardized acquisition of blood sample for Hct after the CMR examination, when the patient is still in supine position, would increase the precision of ECV measurements.
Collapse
Affiliation(s)
- Henrik Engblom
- Department of Clinical Physiology, Clinical Sciences, Lund University and Lund University Hospital, Getingevägen 3, 221 85 Lund, Sweden
| | - Mikael Kanski
- Department of Clinical Physiology, Clinical Sciences, Lund University and Lund University Hospital, Getingevägen 3, 221 85 Lund, Sweden
| | - Sascha Kopic
- Department of Clinical Physiology, Clinical Sciences, Lund University and Lund University Hospital, Getingevägen 3, 221 85 Lund, Sweden
| | - David Nordlund
- Department of Clinical Physiology, Clinical Sciences, Lund University and Lund University Hospital, Getingevägen 3, 221 85 Lund, Sweden
| | - Christos G. Xanthis
- Department of Clinical Physiology, Clinical Sciences, Lund University and Lund University Hospital, Getingevägen 3, 221 85 Lund, Sweden
| | - Robert Jablonowski
- Department of Clinical Physiology, Clinical Sciences, Lund University and Lund University Hospital, Getingevägen 3, 221 85 Lund, Sweden
| | - Einar Heiberg
- Department of Clinical Physiology, Clinical Sciences, Lund University and Lund University Hospital, Getingevägen 3, 221 85 Lund, Sweden
| | - Anthony H. Aletras
- Department of Clinical Physiology, Clinical Sciences, Lund University and Lund University Hospital, Getingevägen 3, 221 85 Lund, Sweden
- Laboratory of Computing, Medical Informatics and Biomedical – Imaging Technologies, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Marcus Carlsson
- Department of Clinical Physiology, Clinical Sciences, Lund University and Lund University Hospital, Getingevägen 3, 221 85 Lund, Sweden
| | - Håkan Arheden
- Department of Clinical Physiology, Clinical Sciences, Lund University and Lund University Hospital, Getingevägen 3, 221 85 Lund, Sweden
| |
Collapse
|
114
|
Schelbert EB, Bering P. Constancy of Spatial Variation in Diffuse Myocardial Disease: Implications for Diagnosing Disease. Circ Cardiovasc Imaging 2018; 11:e007836. [PMID: 29853468 DOI: 10.1161/circimaging.118.007836] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Erik B Schelbert
- UPMC Cardiovascular Magnetic Resonance Center, Heart and Vascular Institute, UMPC, Pittsburgh, PA (E.B.S., P.B.). .,Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, PA (E.B.S.)
| | - Patrick Bering
- UPMC Cardiovascular Magnetic Resonance Center, Heart and Vascular Institute, UMPC, Pittsburgh, PA (E.B.S., P.B.)
| |
Collapse
|
115
|
Fang G, Chen S, Huang Q, Chen L, Liao D. Curcumin suppresses cardiac fibroblasts activities by regulating the proliferation and cell cycle via the inhibition of the p38 MAPK/ERK signaling pathway. Mol Med Rep 2018; 18:1433-1438. [PMID: 29901190 PMCID: PMC6072161 DOI: 10.3892/mmr.2018.9120] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 05/09/2018] [Indexed: 12/12/2022] Open
Abstract
Cardiac fibrosis is a deleterious effect of many cardiovascular diseases. Previous studies have shown that curcumin has exhibited protective effects on cardiovascular diseases. The aim of the present study was to evaluate the effects of curcumin on the activity of human cardiac fibroblasts (CFs) and to elucidate the underlying mechanisms involved. Human CFs were incubated with or without curcumin (20 µmol/l) and transforming growth factor β1 (TGF‑β1; 10 ng/ml), and the expression of α‑smooth muscle actin (α‑SMA), collagen type Iα (COLA)‑1 and COLA3 was evaluated using reverse transcription‑quantitative polymerase chain reaction and western blot analysis. Cell proliferation was evaluated by Cell Counting Kit‑8 analysis, and phases of the cell cycle were studied by flow cytometry. Western blot analysis was performed to evaluate the expression of cyclin‑dependent kinase 1 (CDK1), Cyclin B, phosphorylation (p)‑mothers against decapentaplegic homolog 2/3 (p‑smad2/3), p‑P38, and p‑extracellular regulated protein kinases (ERK). Curcumin significantly reduced mRNA and protein levels of α‑SMA, COLA1, and COLA3 in CFs stimulated with TGF‑β1. However, in the absence of TGF‑β1, curcumin did not have any effects on CFs, suggesting that curcumin inhibited TGF‑β1‑mediated CF activities, including differentiation and collagen deposition. Additionally, curcumin inhibited the proliferation of TGF‑β1‑treated CFs, and promoted G2/M phase cell cycle arrest. Curcumin reduced cell cycle protein expression by inhibiting smad2/3, p38 mitogen‑activated protein kinase, and ERK phosphorylation in TGF‑β1‑treated CFs. Thus, these results indicated that curcumin may be a potential anti‑fibrotic drug to treat cardiac fibrosis.
Collapse
Affiliation(s)
- Guanhua Fang
- Department of Cardiac Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P.R. China
| | - Shaoqin Chen
- Department of Cardiac Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P.R. China
| | - Qiuyu Huang
- Department of Cardiac Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P.R. China
| | - Liangwan Chen
- Department of Cardiac Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P.R. China
| | - Dongshan Liao
- Department of Cardiac Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P.R. China
| |
Collapse
|
116
|
Schelbert EB, Miller CA. Myocardial tissue characteristics undoubtedly differ by gender but not age. Eur Heart J Cardiovasc Imaging 2018; 19:611-612. [PMID: 29648631 DOI: 10.1093/ehjci/jey057] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Erik B Schelbert
- UPMC Cardiovascular Magnetic Resonance Center, Heart and Vascular Institute, Pittsburgh, PA, USA.,Department of Medicine, University of Pittsburgh School of Medicine, 200 Lothrop Street, PUH E E354.2, Pittsburgh, PA 15101, USA
| | - Christopher A Miller
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester M13 9PL, UK.,Manchester University NHS Foundation Trust, Wythenshawe Hospital, Southmoor Road, Wythenshawe, Manchester M23 9LT, UK.,Division of Cell-Matrix Biology & Regenerative Medicine, School of Biology, Faculty of Biology, Medicine & Health, Wellcome Centre for Cell-Matrix Research, Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| |
Collapse
|
117
|
Wang R, Liu X, Schoepf UJ, van Assen M, Alimohamed I, Griffith LP, Luo T, Sun Z, Fan Z, Xu L. Extracellular volume quantitation using dual-energy CT in patients with heart failure: Comparison with 3T cardiac MR. Int J Cardiol 2018; 268:236-240. [PMID: 29804697 DOI: 10.1016/j.ijcard.2018.05.027] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 05/03/2018] [Accepted: 05/09/2018] [Indexed: 12/25/2022]
Abstract
BACKGROUNDS Cardiac magnetic resonance (CMR) T1 mapping and the extracellular volume (ECV) have been developed to quantitative analysis of diffusely abnormal myocardial fibrosis (MF). However, dual-energy CT (DECT) has a potential for calculation of ECV. The aim of this study is to evaluate the feasibility and accuracy of DECT technique in determining the ECV in patients with heart failure, with 3T CMR as the reference. METHODS Thirty-five patients with various reasons of heart failure were enrolled in this study. Both DECT and CMR exams were completed within 24 h. ECVs were calculated, and the relationship between DECT-ECV, CMR-ECV, and other heart function parameters, including left ventricular end systolic and diastolic volume, cardiac output and ejection fraction (LVESV, LVEDV, CO, LVEF), Brain natriuretic peptide (BNP) was determined. All participants gave informed consent, and the study was approved by the institutional review board. RESULTS The median ECVs on DECT and CMR were 33% (95%CI: 32%-36%) and 30% (95%CI: 30% - 32%), respectively. A good correlation between myocardial ECV at DECT and that at CMR (r = 0.945, P < 0.001) was observed. Bland-Altman analysis between DECT and CMR showed a small bias (2.6%), with 95% limits of agreement of -0.4% and 5.6%. Interobserver agreement for ECV at DECT was excellent (ICC = 0.907). Both ECVs, for DECT and CMR, were inversely associated with LVEF and CO. CONCLUSION DECT-based ECV could be an alternative non-invasive imaging tool for myocardial tissue characterization. However, overestimation of the extent of diffuse MF is observed with use of DECT.
Collapse
Affiliation(s)
- Rui Wang
- Department of Radiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, PR China
| | - Xinmin Liu
- Wards 40 of Cardiology Department, Beijing Anzhen Hospital, Capital Medical University, Beijing, PR China
| | - U Joseph Schoepf
- Division of Cardiovascular Imaging, Department of Radiology and Radiological Science, Medical University or South Carolina, Charleston, SC, USA; Division of Cardiology, Department of Internal Medicine, Medical University or South Carolina, Charleston, SC, USA
| | - Marly van Assen
- Division of Cardiovascular Imaging, Department of Radiology and Radiological Science, Medical University or South Carolina, Charleston, SC, USA; Center for Medical Imaging North-East Netherlands (CMI-NEN), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Imtiaz Alimohamed
- Division of Cardiovascular Imaging, Department of Radiology and Radiological Science, Medical University or South Carolina, Charleston, SC, USA
| | - L Parkwood Griffith
- Division of Cardiovascular Imaging, Department of Radiology and Radiological Science, Medical University or South Carolina, Charleston, SC, USA
| | - Taiyang Luo
- Wards 40 of Cardiology Department, Beijing Anzhen Hospital, Capital Medical University, Beijing, PR China
| | - Zhonghua Sun
- Department of Medical Radiation Sciences, Curtin University, Perth, WA 6845, Australia
| | - Zhanming Fan
- Department of Radiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, PR China
| | - Lei Xu
- Department of Radiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, PR China.
| |
Collapse
|
118
|
Kamimura D, Suzuki T, Hall ME, Wang W, Winniford MD, Shah AM, Rodriguez CJ, Butler KR, Mosley TH. Diastolic wall strain is associated with incident heart failure in African Americans: Insights from the atherosclerosis risk in communities study. J Cardiol 2018; 71:477-483. [PMID: 29203080 PMCID: PMC6100746 DOI: 10.1016/j.jjcc.2017.11.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 11/03/2017] [Accepted: 11/06/2017] [Indexed: 12/16/2022]
Abstract
BACKGROUND Increased left ventricular (LV) myocardial stiffness may be associated with impaired LV hemodynamics and incident heart failure (HF). However, an indicator that estimates LV myocardial stiffness easily and non-invasively is lacking. The purpose of this study was to determine whether diastolic wall strain (DWS), an echocardiographic estimator of LV myocardial stiffness, is associated with incident HF in a middle-aged community-based cohort of African Americans. METHODS AND RESULTS We investigated associations between DWS and incident HF among 1528 African Americans (mean age 58.5 years, 66% women) with preserved LV ejection fraction (EF ≥50%) and without a history of cardiovascular disease in the Atherosclerosis Risk in Communities Study. Participants with the smallest DWS quintile (more LV myocardial stiffness) had a higher LV mass index, higher relative wall thickness, and lower arterial compliance than those in the larger four DWS quintiles (p<0.01 for all). Over a mean follow-up of 15.6 years, there were 251 incident HF events (incidence rate: 10.9 per 1000 person-years). After adjustment for traditional risk factors and incident coronary artery disease, both continuous and categorical DWS were independently associated with incident HF (HR 1.21, 95%CI 1.04-1.41 for 0.1 decrease in continuous DWS, p=0.014, HR 1.40, 95%CI 1.05-1.87 for the smallest DWS quintile vs other combined quintiles, p=0.022). CONCLUSIONS DWS was independently associated with an increased risk of incident HF in a community-based cohort of African Americans. DWS could be used as a qualitative estimator of LV myocardial stiffness.
Collapse
Affiliation(s)
- Daisuke Kamimura
- Department of Medicine-Cardiology, University of Mississippi Medical Center, Jackson, MS, USA.
| | - Takeki Suzuki
- Department of Medicine-Cardiology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Michael E Hall
- Department of Medicine-Cardiology, University of Mississippi Medical Center, Jackson, MS, USA; Mississippi Center for Heart Research, Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, USA
| | - Wanmei Wang
- Center of Biostatistics and Bioinformatics, University of Mississippi Medical Center, Jackson, MS, USA
| | - Michael D Winniford
- Department of Medicine-Cardiology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Amil M Shah
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Carlos J Rodriguez
- Department of Epidemiology and Prevention, Wake Forest School of Medicine, Winston-Salem, NC, USA; Department of Internal Medicine - Section on Cardiovascular Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Kenneth R Butler
- Division of Geriatric Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Thomas H Mosley
- Division of Geriatric Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| |
Collapse
|
119
|
Wong TC, McNamara DM. Imaging-Based Surveillance for Graft Rejection Following Heart Transplantation: Ready for Prime Time? JACC Cardiovasc Imaging 2018; 12:1615-1617. [PMID: 29680340 DOI: 10.1016/j.jcmg.2018.04.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 04/02/2018] [Indexed: 10/17/2022]
Affiliation(s)
- Timothy C Wong
- Department of Medicine, Division of Cardiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; Cardiovascular Magnetic Resonance Center, Heart and Vascular Institute, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania.
| | - Dennis M McNamara
- Department of Medicine, Division of Cardiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; Center for Heart Failure Research, Heart and Vascular Institute, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| |
Collapse
|
120
|
Abstract
Heart failure is a growing cardiovascular disease with significant epidemiological, clinical, and societal implications and represents a high unmet need. Strong efforts are currently underway by academic and industrial researchers to develop novel treatments for heart failure. Biomarkers play an important role in patient selection and monitoring in drug trials and in clinical management. The present review gives an overview of the role of available molecular, imaging, and device-derived digital biomarkers in heart failure drug development and highlights capabilities and limitations of biomarker use in this context.
Collapse
|
121
|
Zhang K, Meng X, Wang W, Zheng J, An S, Wang S, Qi Y, Gao C, Tang YD. Prognostic Value of Free Triiodothyronine Level in Patients With Hypertrophic Obstructive Cardiomyopathy. J Clin Endocrinol Metab 2018; 103:1198-1205. [PMID: 29304228 DOI: 10.1210/jc.2017-02386] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 12/28/2017] [Indexed: 01/01/2023]
Abstract
CONTEXT Thyroid hormone acts as a fundamental regulator in cardiovascular homeostasis in pathophysiological conditions. OBJECTIVE This study aims to determine whether thyroid hormone could be an independent predictor of adverse events in patients with hypertrophic obstructive cardiomyopathy (HOCM). DESIGN, PATIENTS, AND OUTCOME MEASURES The original cohort consisted of 965 consecutive patients with HOCM who were admitted to Fuwai Hospital from October 2009 to December 2014, and 756 patients completed thyroid function evaluations. Patients were divided into three groups according to free triiodothyronine (FT3) levels: tertile 1 (<2.81 pg/mL, n = 247), tertile 2 (2.81 to 3.11 pg/mL, n = 250), tertile 3 (3.12 to 4.09 pg/mL, n = 259). RESULTS In correlation analysis, FT3 showed significantly positive correlation with left ventricular ejection fraction (r = 0.109, P = 0.003). After a median follow-up of 44 months, a total of 45 (6.0%) endpoints (all-cause mortality or cardiac transplantation) occurred with rates of 13.4%, 3.6%, and 1.2% in tertiles 1, 2, and 3, respectively. Univariate Cox analysis established FT3 as a predictor of endpoint [hazard ratio (HR), 0.111; 95% confidence interval (CI), 0.065, 0.189; P < 0.001]. After adjustment for traditional risk factors, the prognostic value of FT3 level was still significant (HR, 0.216; 95% CI, 0.083, 0.559; P = 0.002). Compared with patients in tertile 3, those in tertile 1 were at a much higher risk of endpoint (HR, 4.918; 95% CI, 1.076, 22.485; P = 0.040). CONCLUSIONS FT3 correlated with cardiac function and could serve as an independent predictor of all-cause mortality and cardiac transplantation in patients with HOCM. These results suggest that monitoring thyroid function in HOCM patients is necessary.
Collapse
Affiliation(s)
- Kuo Zhang
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiangbin Meng
- Department of Cardiology, Zhengzhou University People's Hospital, Zhengzhou, Henan, China
| | - Wenyao Wang
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jilin Zheng
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shimin An
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Siyuan Wang
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yu Qi
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chuanyu Gao
- Department of Cardiology, Zhengzhou University People's Hospital, Zhengzhou, Henan, China
| | - Yi-Da Tang
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
122
|
Aus dem Siepen F, Baumgärtner C, Müller-Henessen M, André F, Messroghli D, Ochs M, Riffel J, Giannitsis E, Katus HA, Friedrich MG, Buss SJ. Variability of cardiovascular magnetic resonance (CMR) T1 mapping parameters in healthy volunteers during long-term follow-up. Open Heart 2018. [PMID: 29531760 PMCID: PMC5845426 DOI: 10.1136/openhrt-2017-000717] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Background Myocardial T1 and extracellular volume (ECV) derived from cardiovascular MRIs are more and more widely accepted as important markers for diagnosis, risk prediction and monitoring of cardiac disease. Yet data regarding long-term stability of myocardial T1 mapping are lacking. The aim of this study was to investigate the long-term stability of native and postcontrast T1 mapping values in healthy volunteers. Methods 18 strictly selected healthy volunteers (52±10 years, 12 men) were studied on a Philips Achieva 1.5 Tesla scanner. T1 relaxation times were measured before and 15 min after a bolus contrast injection of gadolinium diethylenetriamine penta-acetic acid (DTPA) (0.2 mmol/kg) using a single-breath-hold modified Look-Locker inversion recovery 3(3)3(3)5 sequence. ECV was calculated using native and postcontrast T1 times of myocardium and blood correcting for blood haematocrit. Exams were repeated 3.6±0.5 years later under the same conditions and using the same scan protocols. Results Cardiac biomarkers (high-sensitivity troponin T and N terminal pro-brain natriuretic peptide) remained unchanged, as well as left ventricular mass, and global and longitudinal function. No significant change occurred regarding native T1 times (1017±24 ms vs 1015±21 ms; P=0.6), postcontrast T1 times (426±38 ms vs 413±20 ms; P=0.13) or ECV (22%±2% vs 23%±2%; P=0.3). Native T1 time and ECV appeared to be better reproducible than postcontrast T1, resulting in lower coefficients of variation (ECV: 3.5%, native T1: 1.3%, postcontrast T1: 6.4%) and smaller limits of agreement (ECV: 2%/−2%, native T1: 39 ms/−35 ms, postcontrast T1: 85 ms/−59 ms). Conclusions During long-term follow-up, native T1 and ECV values are very robust markers, whereas postcontrast T1 results appear less stable.
Collapse
Affiliation(s)
- Fabian Aus dem Siepen
- Department of Cardiology, Angiology and Respiratory Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Christian Baumgärtner
- Department of Cardiology, Angiology and Respiratory Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Matthias Müller-Henessen
- Department of Cardiology, Angiology and Respiratory Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Florian André
- Department of Cardiology, Angiology and Respiratory Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Daniel Messroghli
- Department of Internal Medicine - Cardiology, Deutsches Herzzentrum Berlin and Charité, University Medicine Berlin, Berlin, Germany
| | - Marco Ochs
- Department of Cardiology, Angiology and Respiratory Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Johannes Riffel
- Department of Cardiology, Angiology and Respiratory Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Evangelos Giannitsis
- Department of Cardiology, Angiology and Respiratory Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Hugo A Katus
- Department of Cardiology, Angiology and Respiratory Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Matthias G Friedrich
- Department of Cardiology, Angiology and Respiratory Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Sebastian J Buss
- Department of Cardiology, Angiology and Respiratory Medicine, University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
123
|
Vaduganathan M, Patel RB, Shah SJ, Butler J. Sudden cardiac death in heart failure with preserved ejection fraction: a target for therapy? Heart Fail Rev 2018; 21:455-62. [PMID: 26762335 DOI: 10.1007/s10741-016-9525-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
The incidence and mechanisms of sudden cardiac death (SCD) among patients with heart failure with reduced ejection fraction have been well characterized. Conversely, limited data are available exploring the landscape of SCD in patients with heart failure with preserved ejection fraction (HFpEF). HFpEF is a heterogeneous clinical syndrome of increasing prevalence and is associated with substantial morbidity and mortality. This review will aim to contextualize recent data regarding rates and predictors of SCD in this growing population and to discuss the potential role of pharmacologic and device therapy for prevention of SCD within the at-risk HFpEF subset.
Collapse
Affiliation(s)
- Muthiah Vaduganathan
- Brigham and Women's Hospital Heart and Vascular Center and Harvard Medical School, 75 Francis St, Boston, MA, 02115, USA.
| | - Ravi B Patel
- Brigham and Women's Hospital Heart and Vascular Center and Harvard Medical School, 75 Francis St, Boston, MA, 02115, USA
| | - Sanjiv J Shah
- Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Javed Butler
- Division of Cardiology, Stony Brook University, Stony Brook, NY, USA
| |
Collapse
|
124
|
Habib G, Bucciarelli-Ducci C, Caforio A, Cardim N, Charron P, Cosyns B, Dehaene A, Derumeaux G, Donal E, Dweck M, Edvardsen T, Erba P, Ernande L, Gaemperli O, Galderisi M, Grapsa J, Jacquier A, Klingel K, Lancellotti P, Neglia D, Pepe A, Perrone-Filardi P, Petersen S, Plein S, Popescu B, Reant P, Sade LE, Salaun E, Slart R, Tribouilloy C, Zamorano J. Multimodality imaging in restrictive cardiomyopathies: an european association of cardiovascular imaging expert consensus document in collaboration with the “Working group on myocardial and pericardial diseases” of the european society of cardiology endorsed by the indian academy of echocardiography. JOURNAL OF THE INDIAN ACADEMY OF ECHOCARDIOGRAPHY & CARDIOVASCULAR IMAGING 2018. [DOI: 10.4103/2543-1463.227042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
125
|
Liu Y, Qi H, E M, Shi P, Zhang Q, Li S, Wang Y, Cao Y, Chen Y, Ba L, Gao J, Huang W, Sun H. Transient receptor potential vanilloid-3 (TRPV3) activation plays a central role in cardiac fibrosis induced by pressure overload in rats via TGF-β1 pathway. Naunyn Schmiedebergs Arch Pharmacol 2017; 391:131-143. [DOI: 10.1007/s00210-017-1443-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2017] [Accepted: 11/27/2017] [Indexed: 01/01/2023]
|
126
|
Zareba W, Zareba KM. Cardiac Magnetic Resonance in Sudden Cardiac Arrest Survivors. Circ Cardiovasc Imaging 2017; 10:e007290. [PMID: 29237610 DOI: 10.1161/circimaging.117.007290] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 11/21/2017] [Indexed: 01/15/2023]
Affiliation(s)
- Wojciech Zareba
- From the Heart Research Follow-Up Program, Cardiology Division, University of Rochester Medical Center, NY (W.Z.); and Division of Cardiovascular Medicine, The Ohio State University Wexner Medical Center, Columbus (K.M.Z.).
| | - Karolina M Zareba
- From the Heart Research Follow-Up Program, Cardiology Division, University of Rochester Medical Center, NY (W.Z.); and Division of Cardiovascular Medicine, The Ohio State University Wexner Medical Center, Columbus (K.M.Z.)
| |
Collapse
|
127
|
Lottonen-Raikaslehto L, Rissanen R, Gurzeler E, Merentie M, Huusko J, Schneider JE, Liimatainen T, Ylä-Herttuala S. Left ventricular remodeling leads to heart failure in mice with cardiac-specific overexpression of VEGF-B 167: echocardiography and magnetic resonance imaging study. Physiol Rep 2017; 5:5/6/e13096. [PMID: 28351964 PMCID: PMC5371547 DOI: 10.14814/phy2.13096] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 10/07/2016] [Accepted: 11/20/2016] [Indexed: 01/24/2023] Open
Abstract
Cardiac-specific overexpression of vascular endothelial growth factor (VEGF)-B167 is known to induce left ventricular hypertrophy due to altered lipid metabolism, in which ceramides accumulate to the heart and cause mitochondrial damage. The aim of this study was to evaluate and compare different imaging methods to find the most sensitive way to diagnose at early stage the progressive left ventricular remodeling leading to heart failure. Echocardiography and cardiovascular magnetic resonance imaging were compared for imaging the hearts of transgenic mice with cardiac-specific overexpression of VEGF-B167 and wild-type mice from 5 to 14 months of age at several time points. Disease progression was verified by molecular biology methods and histology. We showed that left ventricular remodeling is already ongoing at the age of 5 months in transgenic mice leading to heart failure by the age of 14 months. Measurements from echocardiography and cardiovascular magnetic resonance imaging revealed similar changes in cardiac structure and function in the transgenic mice. Changes in histology, gene expressions, and electrocardiography supported the progression of left ventricular hypertrophy. Longitudinal relaxation time in rotating frame (T1ρ ) in cardiovascular magnetic resonance imaging could be suitable for detecting severe fibrosis in the heart. We conclude that cardiac-specific overexpression of VEGF-B167 leads to left ventricular remodeling at early age and is a suitable model to study heart failure development with different imaging methods.
Collapse
Affiliation(s)
- Line Lottonen-Raikaslehto
- Department of Biotechnology and Molecular Medicine, A. I. Virtanen Institute for Molecular Sciences, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Riina Rissanen
- Department of Biotechnology and Molecular Medicine, A. I. Virtanen Institute for Molecular Sciences, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Erika Gurzeler
- Department of Biotechnology and Molecular Medicine, A. I. Virtanen Institute for Molecular Sciences, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Mari Merentie
- Department of Biotechnology and Molecular Medicine, A. I. Virtanen Institute for Molecular Sciences, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Jenni Huusko
- Department of Biotechnology and Molecular Medicine, A. I. Virtanen Institute for Molecular Sciences, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Jurgen E Schneider
- Radcliffe Department of Medicine, Division of Cardiovascular Medicine, University of Oxford, United kingdom
| | - Timo Liimatainen
- Department of Biotechnology and Molecular Medicine, A. I. Virtanen Institute for Molecular Sciences, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland.,Clinical Imaging Center, Kuopio University Hospital, Kuopio, Finland
| | - Seppo Ylä-Herttuala
- Department of Biotechnology and Molecular Medicine, A. I. Virtanen Institute for Molecular Sciences, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland .,Gene Therapy Unit, Kuopio University Hospital, Kuopio, Finland.,Heart Center, Kuopio University Hospital, Kuopio, Finland
| |
Collapse
|
128
|
Cavalcante JL, Rijal S, Abdelkarim I, Althouse AD, Sharbaugh MS, Fridman Y, Soman P, Forman DE, Schindler JT, Gleason TG, Lee JS, Schelbert EB. Cardiac amyloidosis is prevalent in older patients with aortic stenosis and carries worse prognosis. J Cardiovasc Magn Reson 2017; 19:98. [PMID: 29212513 PMCID: PMC5719789 DOI: 10.1186/s12968-017-0415-x] [Citation(s) in RCA: 140] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 11/23/2017] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Non-invasive cardiac imaging allows detection of cardiac amyloidosis (CA) in patients with aortic stenosis (AS). Our objective was to estimate the prevalence of clinically suspected CA in patients with moderate and severe AS referred for cardiovascular magnetic resonance (CMR) in age and gender categories, and assess associations between AS-CA and all-cause mortality. METHODS We retrospectively identified consecutive AS patients defined by echocardiography referred for further CMR assessment of valvular, myocardial, and aortic disease. CMR identified CA based on typical late-gadolinium enhancement (LGE) patterns, and ancillary clinical evaluation identified suspected CA. Survival analysis with the Log rank test and Cox regression compared associations between CA and mortality. RESULTS There were 113 patients (median age 74 years, Q1-Q3: 62-82 years), 96 (85%) with severe AS. Suspected CA was present in 9 patients (8%) all > 80 years. Among those over the median age of 74 years, the prevalence of CA was 9/57 (16%), and excluding women, the prevalence was 8/25 (32%). Low-flow, low-gradient physiology was very common in CA (7/9 patients or 78%). Over a median follow-up of 18 months, 40 deaths (35%) occurred. Mortality in AS + CA patients was higher than AS alone (56% vs. 20% at 1-year, log rank 15.0, P < 0.0001). Adjusting for aortic valve replacement modeled as a time-dependent covariate, Society of Thoracic Surgery predicted risk of mortality, left ventricular ejection fraction, CA remained associated with all-cause mortality (HR = 2.92, 95% CI = 1.09-7.86, P = 0.03). CONCLUSIONS Suspected CA appears prevalent among older male patients with AS, especially with low flow, low gradient AS, and associates with all-cause mortality. The importance of screening for CA in older AS patients and optimal treatment strategies in those with CA warrant further investigation, especially in the era of transcatheter aortic valve implantation.
Collapse
Affiliation(s)
- João L. Cavalcante
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, 200 Lothrop Street, Scaife Hall S-558, Pittsburgh, PA 15213 USA
- UPMC Cardiovascular Magnetic Resonance Center, Heart and Vascular Institute, Pittsburgh, Pennsylvania, USA
| | - Shasank Rijal
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, 200 Lothrop Street, Scaife Hall S-558, Pittsburgh, PA 15213 USA
| | - Islam Abdelkarim
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, 200 Lothrop Street, Scaife Hall S-558, Pittsburgh, PA 15213 USA
| | - Andrew D. Althouse
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, 200 Lothrop Street, Scaife Hall S-558, Pittsburgh, PA 15213 USA
| | - Michael S. Sharbaugh
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, 200 Lothrop Street, Scaife Hall S-558, Pittsburgh, PA 15213 USA
| | - Yaron Fridman
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, 200 Lothrop Street, Scaife Hall S-558, Pittsburgh, PA 15213 USA
- UPMC Cardiovascular Magnetic Resonance Center, Heart and Vascular Institute, Pittsburgh, Pennsylvania, USA
| | - Prem Soman
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, 200 Lothrop Street, Scaife Hall S-558, Pittsburgh, PA 15213 USA
| | - Daniel E. Forman
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, 200 Lothrop Street, Scaife Hall S-558, Pittsburgh, PA 15213 USA
| | - John T. Schindler
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, 200 Lothrop Street, Scaife Hall S-558, Pittsburgh, PA 15213 USA
| | - Thomas G. Gleason
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, 200 Lothrop Street, Scaife Hall S-558, Pittsburgh, PA 15213 USA
| | - Joon S. Lee
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, 200 Lothrop Street, Scaife Hall S-558, Pittsburgh, PA 15213 USA
| | - Erik B. Schelbert
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, 200 Lothrop Street, Scaife Hall S-558, Pittsburgh, PA 15213 USA
- UPMC Cardiovascular Magnetic Resonance Center, Heart and Vascular Institute, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
129
|
Liu B, Edwards NC, Neal DAH, Weston C, Nash G, Nikolaidis N, Barker T, Patel R, Bhabra M, Steeds RP. A prospective study examining the role of myocardial Fibrosis in outcome following mitral valve repair IN DEgenerative mitral Regurgitation: rationale and design of the mitral FINDER study. BMC Cardiovasc Disord 2017; 17:282. [PMID: 29166877 PMCID: PMC5700678 DOI: 10.1186/s12872-017-0715-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 11/12/2017] [Indexed: 02/04/2023] Open
Abstract
Background The optimal management of chronic severe primary degenerative mitral regurgitation (MR) is to repair the valve but identification of the optimal timing of surgery remains challenging. Current guidelines suggest ‘watchful waiting’ until the onset of symptoms or left ventricular (LV) dysfunction but these have been challenged as promoting ‘rescue surgery’. Better predictors are required to inform decision-making in relation to the necessity and timing of surgery. Chronic volume overload is a stimulus for adverse adaptive LV remodelling. Subclinical reduction in LV strain before mitral repair predicts a fall in LV ejection fraction following surgery and is thought to reflect the development of myocardial fibrosis in response to chronic volume overload. Myocardial fibrosis can be detected non-invasively using cardiac magnetic resonance (CMR) imaging techniques as an expansion of the extracellular volume (ECV). Methods/design This study investigates whether: 1) patients with above median ECV will have smaller reduction in end-systolic volume index (as a measure of the degree of reverse LV remodelling) on CMR following mitral valve repair, compared to those with below median ECV; and 2) higher ECV on CMR, validated through histology, adversely impacts upon post-operative complications and symptomatic improvement following surgery. This is a multi-centre, prospective, cross-sectional comparison of patients prior to and 9 months following surgery for chronic severe primary degenerative MR. To establish the natural history of ECV in MR, an additional cohort of patients with asymptomatic MR who do not wish to consider early repair will be followed. Investigations include CMR, cardiopulmonary exercise test, stress echocardiography, signal-averaged electrocardiogram, 24-h electrocardiogram monitoring, laboratory tests and patient-reported outcome measures. Patients undergoing surgery will have cardiac biopsies performed at the time of mitral valve repair for histological quantification of fibrosis. Discussion This study will advance our understanding of ventricular remodelling in MR, its impact on patient symptoms and ventricular response following surgery. Establishing the link between myocardial fibrosis (measured on CMR and validated through histology), with early ventricular dysfunction, will offer physicians a novel non-invasive biomarker that can further inform the timing of surgery. Trial registration This trial was registered at ClinicalTrials.gov (NCT02355418) on 30th November 2015.
Collapse
Affiliation(s)
- Boyang Liu
- University Hospital Birmingham NHS Foundation Trust, Birmingham, UK. .,Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK.
| | - Nicola C Edwards
- University Hospital Birmingham NHS Foundation Trust, Birmingham, UK.,Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
| | - Desley A H Neal
- University Hospital Birmingham NHS Foundation Trust, Birmingham, UK
| | - Christopher Weston
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
| | - Gerard Nash
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
| | | | - Thomas Barker
- University Hospitals Coventry and Warwickshire NHS Trust, Coventry, UK
| | - Ramesh Patel
- University Hospitals Coventry and Warwickshire NHS Trust, Coventry, UK
| | - Moninder Bhabra
- University Hospital Birmingham NHS Foundation Trust, Birmingham, UK
| | - Richard P Steeds
- University Hospital Birmingham NHS Foundation Trust, Birmingham, UK.,Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
| |
Collapse
|
130
|
Weber KT, Sun Y, Gerling IC, Guntaka RV. Regression of Established Cardiac Fibrosis in Hypertensive Heart Disease. Am J Hypertens 2017; 30:1049-1052. [PMID: 28379281 DOI: 10.1093/ajh/hpx054] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 03/20/2017] [Indexed: 01/19/2023] Open
Abstract
Established cardiac fibrosis (ECF) with symptomatic heart failure preserved ejection fraction represents an ever-increasing segment of the hypertensive population. The regression of ECF with attendant improvement in myocardial stiffness and symptomatic failure represents an unmet health care need. Is the regression of ECF in hypertensive heart disease feasible and will stiffness and symptomatic failure be improved? What is the cellular/molecular signaling involved in its regression? What incremental knowledge is needed to proceed effectively? These issues are addressed in this Review.
Collapse
Affiliation(s)
- Karl T Weber
- Division of Cardiovascular Diseases, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Yao Sun
- Division of Cardiovascular Diseases, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Ivan C Gerling
- Division of Endocrinology, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Ramareddy V Guntaka
- Department of Microbiology, Immunology & Biochemistry, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| |
Collapse
|
131
|
Greene SJ, Mentz RJ. Potential advantages of torsemide in patients with heart failure: more than just a 'water pill'? Eur J Heart Fail 2017; 20:471-473. [PMID: 29082584 DOI: 10.1002/ejhf.1024] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 08/28/2017] [Indexed: 12/28/2022] Open
Affiliation(s)
- Stephen J Greene
- Duke Clinical Research Institute, Durham, NC, USA.,Division of Cardiology, Duke University Medical Center, Durham, NC, USA
| | - Robert J Mentz
- Duke Clinical Research Institute, Durham, NC, USA.,Division of Cardiology, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
132
|
Messroghli DR, Moon JC, Ferreira VM, Grosse-Wortmann L, He T, Kellman P, Mascherbauer J, Nezafat R, Salerno M, Schelbert EB, Taylor AJ, Thompson R, Ugander M, van Heeswijk RB, Friedrich MG. Clinical recommendations for cardiovascular magnetic resonance mapping of T1, T2, T2* and extracellular volume: A consensus statement by the Society for Cardiovascular Magnetic Resonance (SCMR) endorsed by the European Association for Cardiovascular Imaging (EACVI). J Cardiovasc Magn Reson 2017; 19:75. [PMID: 28992817 PMCID: PMC5633041 DOI: 10.1186/s12968-017-0389-8] [Citation(s) in RCA: 1036] [Impact Index Per Article: 148.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2017] [Accepted: 09/25/2017] [Indexed: 12/14/2022] Open
Abstract
Parametric mapping techniques provide a non-invasive tool for quantifying tissue alterations in myocardial disease in those eligible for cardiovascular magnetic resonance (CMR). Parametric mapping with CMR now permits the routine spatial visualization and quantification of changes in myocardial composition based on changes in T1, T2, and T2*(star) relaxation times and extracellular volume (ECV). These changes include specific disease pathways related to mainly intracellular disturbances of the cardiomyocyte (e.g., iron overload, or glycosphingolipid accumulation in Anderson-Fabry disease); extracellular disturbances in the myocardial interstitium (e.g., myocardial fibrosis or cardiac amyloidosis from accumulation of collagen or amyloid proteins, respectively); or both (myocardial edema with increased intracellular and/or extracellular water). Parametric mapping promises improvements in patient care through advances in quantitative diagnostics, inter- and intra-patient comparability, and relatedly improvements in treatment. There is a multitude of technical approaches and potential applications. This document provides a summary of the existing evidence for the clinical value of parametric mapping in the heart as of mid 2017, and gives recommendations for practical use in different clinical scenarios for scientists, clinicians, and CMR manufacturers.
Collapse
Affiliation(s)
- Daniel R. Messroghli
- Department of Internal Medicine and Cardiology, Deutsches Herzzentrum Berlin, Berlin, Germany
- Department of Internal Medicine and Cardiology, Charité Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - James C. Moon
- University College London and Barts Heart Centre, London, UK
| | - Vanessa M. Ferreira
- Oxford Centre for Clinical Magnetic Resonance Research, Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Lars Grosse-Wortmann
- Division of Cardiology in the Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, ON Canada
| | - Taigang He
- Cardiovascular Science Research Centre, St George’s, University of London, London, UK
| | | | - Julia Mascherbauer
- Department of Internal Medicine II, Division of Cardiology, Vienna, Austria
| | - Reza Nezafat
- Department of Medicine (Cardiovascular Division), Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, USA
| | - Michael Salerno
- Departments of Medicine Cardiology Division, Radiology and Medical Imaging, and Biomedical Engineering, University of Virginia Health System, Charlottesville, VA USA
| | - Erik B. Schelbert
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
- UPMC Cardiovascular Magnetic Resonance Center, Heart and Vascular Institute, Pittsburgh, PA USA
- Clinical and Translational Science Institute, University of Pittsburgh, Pittsburgh, PA USA
| | - Andrew J. Taylor
- The Alfred Hospital, Baker Heart and Diabetes Institute, Melbourne, Australia
| | - Richard Thompson
- Department of Biomedical Engineering, University of Alberta, Edmonton, Canada
| | - Martin Ugander
- Department of Clinical Physiology, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Ruud B. van Heeswijk
- Department of Radiology, Lausanne University Hospital (CHUV) and Lausanne University (UNIL), Lausanne, Switzerland
| | - Matthias G. Friedrich
- Departments of Medicine and Diagnostic Radiology, McGill University, Montréal, Québec Canada
- Department of Medicine, Heidelberg University, Heidelberg, Germany
- Département de radiologie, Université de Montréal, Montréal, Québec Canada
- Departments of Cardiac Sciences and Radiology, University of Calgary, Calgary, Canada
| |
Collapse
|
133
|
Marian AJ, Braunwald E. Hypertrophic Cardiomyopathy: Genetics, Pathogenesis, Clinical Manifestations, Diagnosis, and Therapy. Circ Res 2017; 121:749-770. [PMID: 28912181 DOI: 10.1161/circresaha.117.311059] [Citation(s) in RCA: 776] [Impact Index Per Article: 110.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Hypertrophic cardiomyopathy (HCM) is a genetic disorder that is characterized by left ventricular hypertrophy unexplained by secondary causes and a nondilated left ventricle with preserved or increased ejection fraction. It is commonly asymmetrical with the most severe hypertrophy involving the basal interventricular septum. Left ventricular outflow tract obstruction is present at rest in about one third of the patients and can be provoked in another third. The histological features of HCM include myocyte hypertrophy and disarray, as well as interstitial fibrosis. The hypertrophy is also frequently associated with left ventricular diastolic dysfunction. In the majority of patients, HCM has a relatively benign course. However, HCM is also an important cause of sudden cardiac death, particularly in adolescents and young adults. Nonsustained ventricular tachycardia, syncope, a family history of sudden cardiac death, and severe cardiac hypertrophy are major risk factors for sudden cardiac death. This complication can usually be averted by implantation of a cardioverter-defibrillator in appropriate high-risk patients. Atrial fibrillation is also a common complication and is not well tolerated. Mutations in over a dozen genes encoding sarcomere-associated proteins cause HCM. MYH7 and MYBPC3, encoding β-myosin heavy chain and myosin-binding protein C, respectively, are the 2 most common genes involved, together accounting for ≈50% of the HCM families. In ≈40% of HCM patients, the causal genes remain to be identified. Mutations in genes responsible for storage diseases also cause a phenotype resembling HCM (genocopy or phenocopy). The routine applications of genetic testing and preclinical identification of family members represents an important advance. The genetic discoveries have enhanced understanding of the molecular pathogenesis of HCM and have stimulated efforts designed to identify new therapeutic agents.
Collapse
Affiliation(s)
- Ali J Marian
- From the Center for Cardiovascular Genetics, Institute of Molecular Medicine, Department of Medicine, University of Texas Health Sciences Center at Houston (A.J.M.); Texas Heart Institute, Houston (A.J.M.); and TIMI Study Group, Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (E.B.).
| | - Eugene Braunwald
- From the Center for Cardiovascular Genetics, Institute of Molecular Medicine, Department of Medicine, University of Texas Health Sciences Center at Houston (A.J.M.); Texas Heart Institute, Houston (A.J.M.); and TIMI Study Group, Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (E.B.)
| |
Collapse
|
134
|
Valor diagnóstico y pronóstico del mapeo de T 1 mediante RMC de los pacientes con insuficiencia cardiaca y fracción de eyección conservada. Rev Esp Cardiol 2017. [DOI: 10.1016/j.recesp.2016.12.033] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
135
|
Feingold B, Picarsic J, Lesniak A, Popp BA, Wood-Trageser MA, Demetris AJ. Late graft dysfunction after pediatric heart transplantation is associated with fibrosis and microvasculopathy by automated, digital whole-slide analysis. J Heart Lung Transplant 2017; 36:1336-1343. [PMID: 29055602 DOI: 10.1016/j.healun.2017.09.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 08/23/2017] [Accepted: 09/26/2017] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Histopathologic features of late graft dysfunction (LGD) in endomyocardial biopsies (EMBs) after pediatric heart transplantation (HT) have been incompletely described and rarely quantified. We employed automated, morphometric analysis of whole-slide EMB images to objectively quantify fibrosis and microvasculopathy after pediatric HT. METHODS Nine recipients with clinical LGD were matched with controls on age, listing diagnosis, crossmatch and time since HT. Fibrosis was quantified as percent tissue area with fibrosis and capillary density as capillaries per unit area, number of capillary "neighbors" within 30 μm of each myocyte and myocyte-to-nearest-capillary diffusion distance. Clinical data, including all EMB reports, were also reviewed. RESULTS The groups were well matched for age at HT (median 4.0 vs 3.1 years), listing diagnosis (50% congenital heart disease for each), positive crossmatch (11% each) and days post-HT (2,628 vs 2,894, p = 0.69). Despite a similar number of previous EMBs (median 23 each, p = 0.43), areas occupied by fibrosis were greater in LGD cases (44.5% vs 23.2%, p = 0.012). Capillary number/area data were not statistically different between LGD cases and controls (378/mm2 vs 559/mm2, p = 0.57), but LGD cases more commonly had zero capillary neighbors (35% vs 20%, p = 0.02) and greater myocyte-to-nearest-capillary distances (27.1 μm vs 18.7 μm, p = 0.005). Cumulative rejection history correlated with fibrosis (r = 0.49, p = 0.039) and myocyte-to-nearest-capillary distance (r = 0.5, p = 0.036). CONCLUSIONS LGD after pediatric HT is associated with previous rejection and characterized histologically by fibrosis and microvasculopathy, which are not readily appreciated by traditional semi-quantitative EMB analysis. Software-assisted EMB analysis may enable greater pathophysiologic understanding of LGD and identification of targets for future study and intervention.
Collapse
Affiliation(s)
- Brian Feingold
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA; Clinical and Translational Science, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.
| | - Jennifer Picarsic
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Andrew Lesniak
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA; Division of Transplant Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Benjamin A Popp
- Division of Transplant Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Michelle A Wood-Trageser
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA; Division of Transplant Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Anthony J Demetris
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA; Division of Transplant Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
136
|
Raucci FJ, Parra DA, Christensen JT, Hernandez LE, Markham LW, Xu M, Slaughter JC, Soslow JH. Synthetic hematocrit derived from the longitudinal relaxation of blood can lead to clinically significant errors in measurement of extracellular volume fraction in pediatric and young adult patients. J Cardiovasc Magn Reson 2017; 19:58. [PMID: 28768519 PMCID: PMC5541652 DOI: 10.1186/s12968-017-0377-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 07/20/2017] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Extracellular volume fraction (ECV) is altered in pathological cardiac remodeling and predicts death and arrhythmia. ECV can be quantified using cardiovascular magnetic resonance (CMR) T1 mapping but calculation requires a measured hematocrit (Hct). The longitudinal relaxation of blood has been used in adults to generate a synthetic Hct (estimate of true Hct) but has not been validated in pediatric populations. METHODS One hundred fourteen children and young adults underwent a total of 163 CMRs with T1 mapping. The majority of subjects had a measured Hct the same day (N = 146). Native and post-contrast T1 were determined in blood pool, septum, and free wall of mid-LV, avoiding areas of late gadolinium enhancement. Synthetic Hct and ECV were calculated and intraclass correlation coefficient (ICC) and linear regression were used to compare measured and synthetic values. RESULTS The mean age was 16.4 ± 6.4 years and mean left ventricular ejection fraction was 59% ± 9%. The mean measured Hct was 41.8 ± 3.0% compared to the mean synthetic Hct of 43.2% ± 2.9% (p < 0.001, ICC 0.46 [0.27, 0.52]) with the previously published model and 41.8% ± 1.4% (p < 0.001, ICC 0.28 [0.13, 0. 42]) with the locally-derived model. Mean measured mid-free wall ECV was 30.5% ± 4.8% and mean synthetic mid-free wall ECV of local model was 29.7% ± 4.6% (p < 0.001, ICC 0.93 [0.91, 0.95]). Correlations were not affected by heart rate and did not significantly differ in subpopulation analysis. While the ICC was strong, differences between measured and synthetic ECV ranged from -8.4% to 4.3% in the septum and -12.6% to 15.8% in the free wall. Using our laboratory's normal cut-off of 28.5%, 59 patients (37%) were miscategorized (53 false negatives, 6 false positives) with published model ECV. The local model had 37 miscategorizations (20 false negatives, 17 false positives), significantly fewer but still a substantial number (23%). CONCLUSIONS Our data suggest that use of synthetic Hct for the calculation of ECV results in miscategorization of individual patients. This difference may be less significant once synthetic ECV is calculated and averaged over a large research cohort, making it potentially useful as a research tool. However, we recommend formal measurement of Hct in children and young adults for clinical CMRs.
Collapse
Affiliation(s)
- Frank J Raucci
- Department of Pediatrics, Thomas P. Graham Jr, Division of Pediatric Cardiology, Vanderbilt University Medical Center, Nashville, USA.
- Thomas P. Graham, Jr. Division of Pediatric Cardiology, Monroe Carell Jr. Children's Hospital at Vanderbilt, 2200 Children's Way, Suite 5230, Doctors' Office Tower, Nashville, TN, 37232-9119, USA.
| | - David A Parra
- Department of Pediatrics, Thomas P. Graham Jr, Division of Pediatric Cardiology, Vanderbilt University Medical Center, Nashville, USA
- Thomas P. Graham, Jr. Division of Pediatric Cardiology, Monroe Carell Jr. Children's Hospital at Vanderbilt, 2200 Children's Way, Suite 5230, Doctors' Office Tower, Nashville, TN, 37232-9119, USA
| | - Jason T Christensen
- Department of Pediatrics, Thomas P. Graham Jr, Division of Pediatric Cardiology, Vanderbilt University Medical Center, Nashville, USA
- Thomas P. Graham, Jr. Division of Pediatric Cardiology, Monroe Carell Jr. Children's Hospital at Vanderbilt, 2200 Children's Way, Suite 5230, Doctors' Office Tower, Nashville, TN, 37232-9119, USA
| | - Lazaro E Hernandez
- Joe DiMaggio Children's Hospital, Pediatric and Congenital Cardiac MRI program, Los Angeles, CA, USA
| | - Larry W Markham
- Department of Pediatrics, Thomas P. Graham Jr, Division of Pediatric Cardiology, Vanderbilt University Medical Center, Nashville, USA
- Thomas P. Graham, Jr. Division of Pediatric Cardiology, Monroe Carell Jr. Children's Hospital at Vanderbilt, 2200 Children's Way, Suite 5230, Doctors' Office Tower, Nashville, TN, 37232-9119, USA
| | - Meng Xu
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - James C Slaughter
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jonathan H Soslow
- Department of Pediatrics, Thomas P. Graham Jr, Division of Pediatric Cardiology, Vanderbilt University Medical Center, Nashville, USA
- Thomas P. Graham, Jr. Division of Pediatric Cardiology, Monroe Carell Jr. Children's Hospital at Vanderbilt, 2200 Children's Way, Suite 5230, Doctors' Office Tower, Nashville, TN, 37232-9119, USA
| |
Collapse
|
137
|
Vaduganathan M, Patel RB, Michel A, Shah SJ, Senni M, Gheorghiade M, Butler J. Mode of Death in Heart Failure With Preserved Ejection Fraction. J Am Coll Cardiol 2017; 69:556-569. [PMID: 28153111 DOI: 10.1016/j.jacc.2016.10.078] [Citation(s) in RCA: 173] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 10/26/2016] [Indexed: 12/20/2022]
Abstract
Little is known about specific modes of death in patients with heart failure with preserved ejection fraction (HFpEF). Herein, the authors critically appraise the current state of data and offer potential future directions. They conducted a systematic review of 1,608 published HFpEF papers from January 1, 1985, to December 31, 2015, which yielded 8 randomized clinical trials and 24 epidemiological studies with mode-of-death data. Noncardiovascular modes of death represent an important competing risk in HFpEF. Although sudden death accounted for ∼25% to 30% of deaths in trials, its definition is nonspecific; it is unclear what proportion represents arrhythmic deaths. Moving forward, reporting and definitions of modes of death must be standardized and tailored to the HFpEF population. Broad-scale systematic autopsies and long-term rhythm monitoring may clarify the underlying pathology and mechanisms driving mortal events. There is an unmet need for a longitudinal multicenter, global registry of patients with HFpEF to map its natural history.
Collapse
Affiliation(s)
- Muthiah Vaduganathan
- Brigham and Women's Hospital Heart & Vascular Center and Harvard Medical School, Boston, Massachusetts
| | - Ravi B Patel
- Division of Cardiology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | | | - Sanjiv J Shah
- Division of Cardiology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Michele Senni
- Dipartimento Cardiovascolare, Azienda Ospedaliera Papa Giovanni XXIII, Bergamo, Italy
| | - Mihai Gheorghiade
- Center for Cardiovascular Innovation, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Javed Butler
- Division of Cardiology, Stony Brook University, Stony Brook, New York.
| |
Collapse
|
138
|
Hernán Gómez Llambí H, Cao G, Donato M, Suárez D, Ottaviano G, Müller A, Buchholz B, Gelpi R, Otero-Losada M, Milei J. Left ventricular hypertrophy does not prevent heart failure in experimental hypertension. Int J Cardiol 2017; 238:57-65. [PMID: 28410843 DOI: 10.1016/j.ijcard.2017.03.109] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2016] [Revised: 03/04/2017] [Accepted: 03/24/2017] [Indexed: 12/22/2022]
Abstract
BACKGROUND Left ventricular hypertrophy (LVH) secondary to hypertension has been accepted to prevent heart failure (HF) while paradoxically increasing cardiovascular morbi-mortality. OBJECTIVES To evaluate whether antihypertensive treatment inhibits LVH, restores beta-adrenergic response and affects myocardial oxidative metabolism. METHODS Ninety spontaneously hypertensive rats (SHR) were distributed into groups and treated (mg/kg, p.o.) with: losartan 30 (L), hydralazine 11 (H), rosuvastatin 10 (R), carvedilol 20 (C). Hypertension control group comprised 18 normotensive rats (Wistar-Kyoto, WKY). Following euthanasia at 16months, contractility was measured in 50% of rats (Langendorff system) before and after isoproterenol (Iso) 10-9M, 10-7M and 10-5M stimulation. Left ventricular weight (LVW) was measured in the remaining hearts, and normalized by BW. Expression of thioredoxin 1 (Trx-1), peroxyredoxin 2 (Prx-2), glutaredoxin 3 (Grx-3), caspase-3 and brain natriuretic peptide (BNP) was determined. RESULTS Systolic blood pressure (mmHg): 154±3 (L), 137±1 (H), 190±3 (R)*, 206±3 (SHR)*, 183±1 (C)**, and 141±1 (WKY) (*p<0.05 vs. L, H, WKY, **p<0.05 vs. L, H, WKY, SHR). LVW/BW was higher in SHR and R (p<0.05). Groups SHR, R and C evidenced baseline contractile depression. Response to Iso 10-5M was similar in WKY and L. Expression of Trx-1, Prx-2 and Grx-3 increased in C, H, R and L (p<0.01). CONCLUSIONS Present findings argue against the traditional idea and support that LVH might not be required to prevent HF. Increased expression of thioredoxins by antihypertensive treatment might be involved in protection from HF.
Collapse
Affiliation(s)
- H Hernán Gómez Llambí
- Institute of Cardiological Research, School of Medicine, University of Buenos Aires, UBA-CONICET, Buenos Aires, Argentina
| | - G Cao
- Institute of Cardiological Research, School of Medicine, University of Buenos Aires, UBA-CONICET, Buenos Aires, Argentina
| | - M Donato
- Institute of Cardiovascular Pathophysiology, School of Medicine, University of Buenos Aires, UBA-CONICET, Buenos Aires, Argentina
| | - D Suárez
- Institute of Cardiological Research, School of Medicine, University of Buenos Aires, UBA-CONICET, Buenos Aires, Argentina
| | - G Ottaviano
- Institute of Cardiological Research, School of Medicine, University of Buenos Aires, UBA-CONICET, Buenos Aires, Argentina
| | - A Müller
- Institute of Cardiological Research, School of Medicine, University of Buenos Aires, UBA-CONICET, Buenos Aires, Argentina
| | - B Buchholz
- Institute of Cardiovascular Pathophysiology, School of Medicine, University of Buenos Aires, UBA-CONICET, Buenos Aires, Argentina
| | - R Gelpi
- Institute of Cardiovascular Pathophysiology, School of Medicine, University of Buenos Aires, UBA-CONICET, Buenos Aires, Argentina
| | - M Otero-Losada
- Institute of Cardiological Research, School of Medicine, University of Buenos Aires, UBA-CONICET, Buenos Aires, Argentina.
| | - J Milei
- Institute of Cardiological Research, School of Medicine, University of Buenos Aires, UBA-CONICET, Buenos Aires, Argentina
| |
Collapse
|
139
|
Schelbert EB, Sabbah HN, Butler J, Gheorghiade M. Employing Extracellular Volume Cardiovascular Magnetic Resonance Measures of Myocardial Fibrosis to Foster Novel Therapeutics. Circ Cardiovasc Imaging 2017; 10:CIRCIMAGING.116.005619. [PMID: 28512159 DOI: 10.1161/circimaging.116.005619] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Quantifying myocardial fibrosis (MF) with myocardial extracellular volume measures acquired during cardiovascular magnetic resonance promises to transform clinical care by advancing pathophysiologic understanding and fostering novel therapeutics. Extracellular volume quantifies MF by measuring the extracellular compartment depicted by the myocardial uptake of contrast relative to plasma. MF is a key domain of dysfunctional but viable myocardium among others (eg, microvascular dysfunction and cardiomyocyte/mitochondrial dysfunction). Although anatomically distinct, these domains may functionally interact. MF represents pathological remodeling in the heart associated with cardiac dysfunction and adverse outcomes likely mediated by interactions with the microvasculature and the cardiomyocyte. Reversal of MF improves key measures of cardiac dysfunction, so reversal of MF represents a likely mechanism for improved outcomes. Instead of characterizing the myocardium as homogenous tissue and using important yet still generic descriptors, such as thickness (hypertrophy) and function (diastolic or systolic), which lack mechanistic specificity, paradigms of cardiac disease have evolved to conceptualize myocardial disease and patient vulnerability based on the extent of disease involving its various compartments. Specifying myocardial compartmental involvement may then implicate cellular/molecular disease pathways for treatment and targeted pharmaceutical development and above all highlight the role of the cardiac-specific pathology in heart failure among myriad other changes in the heart and beyond. The cardiology community now requires phase 2 and 3 clinical trials to examine strategies for the regression/prevention of MF and eventually biomarkers to identify MF without reliance on cardiovascular magnetic resonance. It seems likely that efficacious antifibrotic therapy will improve outcomes, but definitive data are needed.
Collapse
Affiliation(s)
- Erik B Schelbert
- From the Department of Medicine, University of Pittsburgh School of Medicine, PA (E.B.S.); UPMC Cardiovascular Magnetic Resonance Center, Heart and Vascular Institute, Pittsburgh, PA (E.B.S.); Clinical and Translational Science Institute, University of Pittsburgh, PA (E.B.S.); Division of Cardiovascular Medicine, Department of Medicine, Henry Ford Health System, Detroit, MI (H.N.S.); Cardiology Division, Department of Medicine, Stony Brook University, NY (J.B.); and Center for Cardiovascular Innovation, Northwestern University Feinberg School of Medicine, Chicago, IL (M.G.).
| | - Hani N Sabbah
- From the Department of Medicine, University of Pittsburgh School of Medicine, PA (E.B.S.); UPMC Cardiovascular Magnetic Resonance Center, Heart and Vascular Institute, Pittsburgh, PA (E.B.S.); Clinical and Translational Science Institute, University of Pittsburgh, PA (E.B.S.); Division of Cardiovascular Medicine, Department of Medicine, Henry Ford Health System, Detroit, MI (H.N.S.); Cardiology Division, Department of Medicine, Stony Brook University, NY (J.B.); and Center for Cardiovascular Innovation, Northwestern University Feinberg School of Medicine, Chicago, IL (M.G.)
| | - Javed Butler
- From the Department of Medicine, University of Pittsburgh School of Medicine, PA (E.B.S.); UPMC Cardiovascular Magnetic Resonance Center, Heart and Vascular Institute, Pittsburgh, PA (E.B.S.); Clinical and Translational Science Institute, University of Pittsburgh, PA (E.B.S.); Division of Cardiovascular Medicine, Department of Medicine, Henry Ford Health System, Detroit, MI (H.N.S.); Cardiology Division, Department of Medicine, Stony Brook University, NY (J.B.); and Center for Cardiovascular Innovation, Northwestern University Feinberg School of Medicine, Chicago, IL (M.G.)
| | - Mihai Gheorghiade
- From the Department of Medicine, University of Pittsburgh School of Medicine, PA (E.B.S.); UPMC Cardiovascular Magnetic Resonance Center, Heart and Vascular Institute, Pittsburgh, PA (E.B.S.); Clinical and Translational Science Institute, University of Pittsburgh, PA (E.B.S.); Division of Cardiovascular Medicine, Department of Medicine, Henry Ford Health System, Detroit, MI (H.N.S.); Cardiology Division, Department of Medicine, Stony Brook University, NY (J.B.); and Center for Cardiovascular Innovation, Northwestern University Feinberg School of Medicine, Chicago, IL (M.G.)
| |
Collapse
|
140
|
Prediction of the estimated 5-year risk of sudden cardiac death and syncope or non-sustained ventricular tachycardia in patients with hypertrophic cardiomyopathy using late gadolinium enhancement and extracellular volume CMR. Eur Radiol 2017; 27:5136-5145. [PMID: 28616729 DOI: 10.1007/s00330-017-4869-x] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 03/28/2017] [Accepted: 04/27/2017] [Indexed: 01/23/2023]
Abstract
OBJECTIVES To evaluate the ability of late gadolinium enhancement (LGE) and mapping cardiac magnetic resonance (CMR) including native T1 and global extracellular volume (ECV) to identify hypertrophic cardiomyopathy (HCM) patients at risk for sudden cardiac death (SCD) and to predict syncope or non-sustained ventricular tachycardia (VT). METHODS A 1.5-T CMR was performed in 73 HCM patients and 16 controls. LGE size was quantified using the 3SD, 5SD and full width at half maximum (FWHM) method. T1 and ECV maps were generated by a 3(3)5 modified Look-Locker inversion recovery sequence. Receiver-operating curve analysis evaluated the best parameter to identify patients with increased SCD risk ≥4% and patients with syncope or non-sustained VT. RESULTS Global ECV was the best predictor of SCD risk with an area under the curve (AUC) of 0.83. LGE size was significantly inferior to global ECV with an AUC of 0.68, 0.70 and 0.70 (all P < 0.05) for 3SD-, 5SD- and FWHM-LGE, respectively. Combined use of the SCD risk score and global ECV significantly improved the diagnostic accuracy to identify HCM patients with syncope or non-sustained VT. CONCLUSIONS Combined use of the SCD risk score and global ECV has the potential to improve HCM patient selection, benefiting most implantable cardioverter defibrillators. KEY POINTS • Global ECV identified the best HCM patients with increased SCD risk. • Global ECV performed equally well compared to a SCD risk score. • Combined use of the SCD risk score and global ECV improved test accuracy. • Combined use potentially improves selection of HCM patients for ICD implantation.
Collapse
|
141
|
Towards accurate and precise T 1 and extracellular volume mapping in the myocardium: a guide to current pitfalls and their solutions. MAGNETIC RESONANCE MATERIALS IN PHYSICS BIOLOGY AND MEDICINE 2017; 31:143-163. [PMID: 28608328 PMCID: PMC5813078 DOI: 10.1007/s10334-017-0631-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 05/05/2017] [Accepted: 05/24/2017] [Indexed: 01/27/2023]
Abstract
Mapping of the longitudinal relaxation time (T1) and extracellular volume (ECV) offers a means of identifying pathological changes in myocardial tissue, including diffuse changes that may be invisible to existing T1-weighted methods. This technique has recently shown strong clinical utility for pathologies such as Anderson-Fabry disease and amyloidosis and has generated clinical interest as a possible means of detecting small changes in diffuse fibrosis; however, scatter in T1 and ECV estimates offers challenges for detecting these changes, and bias limits comparisons between sites and vendors. There are several technical and physiological pitfalls that influence the accuracy (bias) and precision (repeatability) of T1 and ECV mapping methods. The goal of this review is to describe the most significant of these, and detail current solutions, in order to aid scientists and clinicians to maximise the utility of T1 mapping in their clinical or research setting. A detailed summary of technical and physiological factors, issues relating to contrast agents, and specific disease-related issues is provided, along with some considerations on the future directions of the field.
Collapse
|
142
|
Ghonim S, Voges I, Gatehouse PD, Keegan J, Gatzoulis MA, Kilner PJ, Babu-Narayan SV. Myocardial Architecture, Mechanics, and Fibrosis in Congenital Heart Disease. Front Cardiovasc Med 2017; 4:30. [PMID: 28589126 PMCID: PMC5440586 DOI: 10.3389/fcvm.2017.00030] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 04/28/2017] [Indexed: 01/15/2023] Open
Abstract
Congenital heart disease (CHD) is the most common category of birth defect, affecting 1% of the population and requiring cardiovascular surgery in the first months of life in many patients. Due to advances in congenital cardiovascular surgery and patient management, most children with CHD now survive into adulthood. However, residual and postoperative defects are common resulting in abnormal hemodynamics, which may interact further with scar formation related to surgical procedures. Cardiovascular magnetic resonance (CMR) has become an important diagnostic imaging modality in the long-term management of CHD patients. It is the gold standard technique to assess ventricular volumes and systolic function. Besides this, advanced CMR techniques allow the acquisition of more detailed information about myocardial architecture, ventricular mechanics, and fibrosis. The left ventricle (LV) and right ventricle have unique myocardial architecture that underpins their mechanics; however, this becomes disorganized under conditions of volume and pressure overload. CMR diffusion tensor imaging is able to interrogate non-invasively the principal alignments of microstructures in the left ventricular wall. Myocardial tissue tagging (displacement encoding using stimulated echoes) and feature tracking are CMR techniques that can be used to examine the deformation and strain of the myocardium in CHD, whereas 3D feature tracking can assess the twisting motion of the LV chamber. Late gadolinium enhancement imaging and more recently T1 mapping can help in detecting fibrotic myocardial changes and evolve our understanding of the pathophysiology of CHD patients. This review not only gives an overview about available or emerging CMR techniques for assessing myocardial mechanics and fibrosis but it also describes their clinical value and how they can be used to detect abnormalities in myocardial architecture and mechanics in CHD patients.
Collapse
Affiliation(s)
- Sarah Ghonim
- Adult Congenital Heart Unit, Royal Brompton Hospital, London, UK
- Cardiovascular Magnetic Resonance Unit, Royal Brompton Hospital, London, UK
- National Heart and Lung Institute, Imperial College, London, UK
| | - Inga Voges
- Adult Congenital Heart Unit, Royal Brompton Hospital, London, UK
- Cardiovascular Magnetic Resonance Unit, Royal Brompton Hospital, London, UK
| | - Peter D. Gatehouse
- Cardiovascular Magnetic Resonance Unit, Royal Brompton Hospital, London, UK
| | - Jennifer Keegan
- Cardiovascular Magnetic Resonance Unit, Royal Brompton Hospital, London, UK
| | - Michael A. Gatzoulis
- Adult Congenital Heart Unit, Royal Brompton Hospital, London, UK
- National Heart and Lung Institute, Imperial College, London, UK
| | - Philip J. Kilner
- Cardiovascular Magnetic Resonance Unit, Royal Brompton Hospital, London, UK
| | - Sonya V. Babu-Narayan
- Adult Congenital Heart Unit, Royal Brompton Hospital, London, UK
- Cardiovascular Magnetic Resonance Unit, Royal Brompton Hospital, London, UK
- National Heart and Lung Institute, Imperial College, London, UK
| |
Collapse
|
143
|
Association between diffuse myocardial fibrosis and diastolic dysfunction in sickle cell anemia. Blood 2017; 130:205-213. [PMID: 28507082 DOI: 10.1182/blood-2017-02-767624] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 05/11/2017] [Indexed: 12/11/2022] Open
Abstract
Sickle cell anemia (SCA)-related cardiomyopathy is characterized by diastolic dysfunction and hyperdynamic features. Diastolic dysfunction portends early mortality in SCA. Diastolic dysfunction is associated with microscopic myocardial fibrosis in SCA mice, but the cause of diastolic dysfunction in humans with SCA is unknown. We used cardiac magnetic resonance measurements of extracellular volume fraction (ECV) to discover and quantify diffuse myocardial fibrosis in 25 individuals with SCA (mean age, 23 ± 13 years) and determine the association between diffuse myocardial fibrosis and diastolic dysfunction. ECV was calculated from pre- and post-gadolinium T1 measurements of blood and myocardium, and diastolic function was assessed by echocardiography. ECV was markedly increased in all participants compared with controls (0.44 ± 0.08 vs 0.26 ± 0.02, P < .0001), indicating the presence of diffuse myocardial fibrosis. Seventeen patients (71%) had diastolic abnormalities, and 7 patients (29%) met the definition of diastolic dysfunction. Participants with diastolic dysfunction had higher ECV (0.49 ± 0.07 vs 0.37 ± 0.04, P = .01) and N-terminal pro-brain natriuretic peptide (NT-proBNP; 191 ± 261 vs 33 ± 33 pg/mL, P = .04) but lower hemoglobin (8.4 ± 0.3 vs 10.9 ± 1.4 g/dL, P = .004) compared with participants with normal diastolic function. Participants with the highest ECV values (≥0.40) were more likely to have diastolic dysfunction (P = .003) and increased left atrial volume (57 ± 11 vs 46 ± 12 mL/m2, P = .04) compared with those with ECV <0.4. ECV correlated with hemoglobin (r = -0.46, P = .03) and NT-proBNP (r = 0.62, P = .001). In conclusion, diffuse myocardial fibrosis, determined by ECV, is a common and previously underappreciated feature of SCA that is associated with diastolic dysfunction, anemia, and high NT-proBNP. Diffuse myocardial fibrosis is a novel mechanism that appears to underlie diastolic dysfunction in SCA.
Collapse
|
144
|
Puntmann VO, Peker E, Chandrashekhar Y, Nagel E. T1 Mapping in Characterizing Myocardial Disease: A Comprehensive Review. Circ Res 2017; 119:277-99. [PMID: 27390332 DOI: 10.1161/circresaha.116.307974] [Citation(s) in RCA: 237] [Impact Index Per Article: 33.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2016] [Accepted: 05/20/2016] [Indexed: 01/06/2023]
Abstract
Cardiovascular magnetic resonance provides insights into myocardial structure and function noninvasively, with high diagnostic accuracy and without ionizing radiation. Myocardial tissue characterization in particular gives cardiovascular magnetic resonance a prime role among all the noninvasive cardiovascular investigations. Late gadolinium enhancement imaging is an established method for visualizing replacement scar, providing diagnostic and prognostic information in a variety of cardiac conditions. Late gadolinium enhancement, however, relies on the regional segregation of tissue characteristics to generate the imaging contrast. Thus, myocardial pathology that is diffuse in nature and affecting the myocardium in a rather uniform and global distribution is not well visualized with late gadolinium enhancement. Examples include diffuse myocardial inflammation, fibrosis, hypertrophy, and infiltration. T1 mapping is a novel technique allowing to diagnose these diffuse conditions by measurement of T1 values, which directly correspond to variation in intrinsic myocardial tissue properties. In addition to providing clinically meaningful indices, T1-mapping measurements also allow for an estimation of extracellular space by calculation of extracellular volume fraction. Multiple lines of evidence suggest a central role for T1 mapping in detection of diffuse myocardial disease in early disease stages and complements late gadolinium enhancement in visualization of the regional changes in common advanced myocardial disease. As a quantifiable measure, it may allow grading of disease activity, monitoring progress, and guiding treatment, potentially as a fast contrast-free clinical application. We present an overview of clinically relevant technical aspects of acquisition and processing, and the current state of art and evidence, supporting its clinical use.
Collapse
Affiliation(s)
- Valentina O Puntmann
- From the Institute for Experimental and Translational Cardiovascular Imaging, DZHK Centre for Cardiovascular Imaging (V.O.P., E.P., E.N.) and Department of Cardiology (V.O.P., E.N.), Goethe University Hospital Frankfurt, Frankfurt am Main, Germany; Department of Radiology, Ankara University School of Medicine, Ankara, Turkey (E.P.); and University of Minnesota and VA Medical Centre, Minneapolis (Y.C.)
| | - Elif Peker
- From the Institute for Experimental and Translational Cardiovascular Imaging, DZHK Centre for Cardiovascular Imaging (V.O.P., E.P., E.N.) and Department of Cardiology (V.O.P., E.N.), Goethe University Hospital Frankfurt, Frankfurt am Main, Germany; Department of Radiology, Ankara University School of Medicine, Ankara, Turkey (E.P.); and University of Minnesota and VA Medical Centre, Minneapolis (Y.C.)
| | - Y Chandrashekhar
- From the Institute for Experimental and Translational Cardiovascular Imaging, DZHK Centre for Cardiovascular Imaging (V.O.P., E.P., E.N.) and Department of Cardiology (V.O.P., E.N.), Goethe University Hospital Frankfurt, Frankfurt am Main, Germany; Department of Radiology, Ankara University School of Medicine, Ankara, Turkey (E.P.); and University of Minnesota and VA Medical Centre, Minneapolis (Y.C.)
| | - Eike Nagel
- From the Institute for Experimental and Translational Cardiovascular Imaging, DZHK Centre for Cardiovascular Imaging (V.O.P., E.P., E.N.) and Department of Cardiology (V.O.P., E.N.), Goethe University Hospital Frankfurt, Frankfurt am Main, Germany; Department of Radiology, Ankara University School of Medicine, Ankara, Turkey (E.P.); and University of Minnesota and VA Medical Centre, Minneapolis (Y.C.).
| |
Collapse
|
145
|
Paiman EHM, Lamb HJ. When should we use contrast material in cardiac MRI? J Magn Reson Imaging 2017; 46:1551-1572. [PMID: 28480596 DOI: 10.1002/jmri.25754] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 04/18/2017] [Indexed: 12/29/2022] Open
Abstract
At present, most of the cardiac magnetic resonance imaging (MRI) examinations rely on contrast-enhanced protocols, but noncontrast alternatives are emerging. Late gadolinium enhancement (LGE) imaging for the detection of myocardial scar can be considered the main cause for the embedding of cardiac MRI into the clinical routine. The novel noncontrast technique of native T1 mapping shows promise for tissue characterization in ischemic and nonischemic cardiomyopathy and may provide additional information over conventional LGE imaging. Technical issues, including measurements variability, still need to be resolved to facilitate a wide clinical application. Ischemia detection can be performed with contrast-based stress perfusion and contrast-free stress wall motion imaging. For coronary magnetic resonance angiography (MRA), protocols with and without contrast material have been developed. Research on coronary atherosclerotic plaque characterization has introduced new applications of contrast material. For MRA of the aorta, which traditionally relied on contrast administration, several noncontrast protocols have become available. This review provides an overview of when to use contrast material in cardiac and cardiac-related vascular MRI, summarizes the major imaging building blocks, and describes the diagnostic value of the available contrast-enhanced and noncontrast techniques. Contrast material in cardiac MRI should be used for LGE imaging for tissue characterization in ischemic or nonischemic cardiomyopathy and may be used for stress perfusion imaging for the detection of ischemia. In cardiac-related vascular MRI, use of contrast material should be avoided, unless high-quality angiography is required that cannot be obtained with noncontrast protocols. LEVEL OF EVIDENCE 5 Technical Efficacy: Stage 3 J. Magn. Reson. Imaging 2017;46:1551-1572.
Collapse
Affiliation(s)
- Elisabeth H M Paiman
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Hildo J Lamb
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
146
|
Youn JC, Hong YJ, Lee HJ, Han K, Shim CY, Hong GR, Suh YJ, Hur J, Kim YJ, Choi BW, Kang SM. Contrast-enhanced T1 mapping-based extracellular volume fraction independently predicts clinical outcome in patients with non-ischemic dilated cardiomyopathy: a prospective cohort study. Eur Radiol 2017; 27:3924-3933. [PMID: 28439651 DOI: 10.1007/s00330-017-4817-9] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Revised: 03/03/2017] [Accepted: 03/17/2017] [Indexed: 01/08/2023]
Abstract
OBJECTIVES We aimed to evaluate the prognostic role of cardiac magnetic resonance imaging (CMR)-based extracellular volume fraction (ECV) in patients with non-ischemic dilated cardiomyopathy (NIDCM) and compare it with late gadolinium enhancement (LGE) parameters. METHODS This was a single-center, prospective, cohort study of 117 NIDCM patients (71 men, 51.9 ± 16.7 years) who underwent clinical 3.0-T CMR. Myocardial ECV and LGE were quantified on the left ventricular myocardium. The presence of midwall LGE was also detected. Nineteen healthy subjects served as controls. The primary end points were cardiovascular (CV) events defined by CV death, rehospitalization due to heart failure, and heart transplantation. RESULTS During the follow-up period (median duration, 11.2 months; 25th-75th percentile, 7.8-21.9 months), the primary end points occurred in 19 patients (16.2%). The ECV (per 3% and 1% increase) was associated with a hazard ratio of 1.80 and 1.22 (95% confidence interval [CI], 1.48-2.20 and 1.14-1.30, respectively; p < 0.001) for the CV events. Multivariable analysis also indicated that ECV was an independent prognostic factor and had a higher prognostic value (Harrell's c statistic, 0.88) than LGE quantification values (0.77) or midwall LGE (0.80). CONCLUSION CMR-based ECV independently predicts the clinical outcome in NIDCM patients. KEY POINTS • T1-mapping-based ECV is a useful parameter of risk stratification in NIDCM • ECV has a higher prognostic value than LGE • Contrast-enhanced T1-mapping CMR is a feasible and safe method.
Collapse
Affiliation(s)
- Jong-Chan Youn
- Division of Cardiology, Dongtan Sacred Heart Hospital, Hallym University College of Medicine, Hwaseong, Republic of Korea
- Division of Cardiology, Severance Cardiovascular Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yoo Jin Hong
- Department of Radiology, Research Institute of Radiological Science, Severance Hospital, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea.
| | - Hye-Jeong Lee
- Department of Radiology, Research Institute of Radiological Science, Severance Hospital, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Kyunghwa Han
- Department of Radiology, Research Institute of Radiological Science, Severance Hospital, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Chi Young Shim
- Division of Cardiology, Severance Cardiovascular Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Geu-Ru Hong
- Division of Cardiology, Severance Cardiovascular Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Young Joo Suh
- Department of Radiology, Research Institute of Radiological Science, Severance Hospital, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Jin Hur
- Department of Radiology, Research Institute of Radiological Science, Severance Hospital, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Young Jin Kim
- Department of Radiology, Research Institute of Radiological Science, Severance Hospital, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Byoung Wook Choi
- Department of Radiology, Research Institute of Radiological Science, Severance Hospital, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Seok-Min Kang
- Division of Cardiology, Severance Cardiovascular Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
147
|
Schelbert EB, Elkayam U, Cooper LT, Givertz MM, Alexis JD, Briller J, Felker GM, Chaparro S, Kealey A, Pisarcik J, Fett JD, McNamara DM. Myocardial Damage Detected by Late Gadolinium Enhancement Cardiac Magnetic Resonance Is Uncommon in Peripartum Cardiomyopathy. J Am Heart Assoc 2017; 6:JAHA.117.005472. [PMID: 28373243 PMCID: PMC5533034 DOI: 10.1161/jaha.117.005472] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
BACKGROUND In peripartum cardiomyopathy, the prevalence of focal myocardial damage detected by late gadolinium enhancement (LGE) cardiovascular magnetic resonance is important to elucidate mechanisms of myocardial injury and cardiac dysfunction. LGE equates irreversible myocardial injury, but LGE prevalence in peripartum cardiomyopathy is uncertain. METHODS AND RESULTS Among 100 women enrolled within the Investigations of Pregnancy Associated Cardiomyopathy cohort, we recruited 40 women at 13 centers to undergo LGE cardiovascular magnetic resonance, enrolled within the first 13 weeks postpartum. Follow-up scans occurred at 6 months postpartum, and death/transplant rates at 12 months. Baseline characteristics did not differ significantly in the parent cohort according to cardiovascular magnetic resonance enrollment except for mechanical circulatory support. LGE was noted only in 2 women (5%) at baseline. While left ventricular dysfunction with enlargement was prevalent at baseline cardiovascular magnetic resonance scans (eg, ejection fraction 38% [Q1-Q3 31-50%], end diastolic volume index=108 mL/m2 [Q1-Q3 83-134 mL/m2]), most women demonstrated significant improvements at 6 months, consistent with a low prevalence of LGE. LGE was not related to baseline clinical variables, ejection fraction, New York Heart Association heart failure class, or mortality. Neither of the 2 women who died exhibited LGE. LGE was inversely associated with persistent left ventricular ejection fraction at 6 months (P=0.006). CONCLUSIONS Factors other than focal myocardial damage detectable by LGE explain the initial transient depressions in baseline left ventricular ejection fraction, yet focal myocardial damage may contribute to persistent myocardial dysfunction and hinder recovery in a small minority. Most women exhibit favorable changes in ventricular function over 6 months. CLINICAL TRIAL REGISTRATION URL: http://www.clinicaltrials.gov. Unique identifier: NCT01085955.
Collapse
Affiliation(s)
| | - Uri Elkayam
- University of Southern California, Los Angeles, CA
| | | | | | - Jeffrey D Alexis
- University of Rochester School of Medicine and Dentistry, Rochester, NY
| | | | | | | | | | | | - James D Fett
- University of Pittsburgh Medical Center, Pittsburgh, PA
| | | | | |
Collapse
|
148
|
Sabbah HN. Silent disease progression in clinically stable heart failure. Eur J Heart Fail 2017; 19:469-478. [PMID: 27976514 PMCID: PMC5396296 DOI: 10.1002/ejhf.705] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Revised: 10/21/2016] [Accepted: 11/02/2016] [Indexed: 12/11/2022] Open
Abstract
Heart failure with reduced ejection fraction (HFrEF) is a progressive disorder whereby cardiac structure and function continue to deteriorate, often despite the absence of clinically apparent signs and symptoms of a worsening disease state. This silent yet progressive nature of HFrEF can contribute to the increased risk of death-even in patients who are 'clinically stable', or who are asymptomatic or only mildly symptomatic-because it often goes undetected and/or undertreated. Current therapies are aimed at improving clinical symptoms, and several agents more directly target the underlying causes of disease; however, new therapies are needed that can more fully address factors responsible for underlying progressive cardiac dysfunction. In this review, mechanisms that drive HFrEF, including ongoing cardiomyocyte loss, mitochondrial abnormalities, impaired calcium cycling, elevated LV wall stress, reactive interstitial fibrosis, and cardiomyocyte hypertrophy, are discussed. Additionally, limitations of current HF therapies are reviewed, with a focus on how these therapies are designed to counteract the deleterious effects of compensatory neurohumoral activation but do not fully prevent disease progression. Finally, new investigational therapies that may improve the underlying molecular, cellular, and structural abnormalities associated with HF progression are reviewed.
Collapse
|
149
|
Diagnostic and Prognostic Value of CMR T 1-Mapping in Patients With Heart Failure and Preserved Ejection Fraction. ACTA ACUST UNITED AC 2017; 70:848-855. [PMID: 28314659 DOI: 10.1016/j.rec.2017.02.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 01/03/2017] [Indexed: 01/09/2023]
Abstract
Heart failure with preserved ejection fraction (HFpEF) presents a major challenge in modern cardiology. Although this syndrome is of increasing prevalence and is associated with unfavorable outcomes, treatment trials have failed to establish effective therapies. Currently, solutions to this dilemma are being investigated, including categorizing and characterizing patients more diversely to individualize treatment. In this regard, new imaging techniques might provide important information. Diastolic dysfunction is a diagnostic and pathophysiological cornerstone in HFpEF and is believed to be caused by systemic inflammation with the development of interstitial myocardial fibrosis and myocardial stiffening. Cardiac magnetic resonance (CMR) T1-mapping is a novel tool, which allows noninvasive quantification of the extracellular space and diffuse myocardial fibrosis. This review provides an overview of the potential of myocardial tissue characterization with CMR T1 mapping in HFpEF patients, outlining its diagnostic and prognostic implications and discussing future directions. We conclude that CMR T1 mapping is potentially an effective tool for patient characterization in large-scale epidemiological, diagnostic, and therapeutic HFpEF trials beyond traditional imaging parameters.
Collapse
|
150
|
Ide S, Riesenkampff E, Chiasson DA, Dipchand AI, Kantor PF, Chaturvedi RR, Yoo SJ, Grosse-Wortmann L. Histological validation of cardiovascular magnetic resonance T1 mapping markers of myocardial fibrosis in paediatric heart transplant recipients. J Cardiovasc Magn Reson 2017; 19:10. [PMID: 28143545 PMCID: PMC5286863 DOI: 10.1186/s12968-017-0326-x] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2016] [Accepted: 01/13/2017] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND Adverse fibrotic remodeling is detrimental to myocardial health and a reliable method for monitoring the development of fibrotic remodeling may be desirable during the follow-up of patients after heart transplantation (HTx). Quantification of diffuse myocardial fibrosis with cardiovascular magnetic resonance (CMR) has been increasingly applied and validated histologically in adult patients with heart disease. However, comparisons of CMR findings with histological fibrosis burden in children are lacking. This study aimed to compare native T1 times and extracellular volume fraction (ECV) derived from CMR with the degree of collagen on endomyocardial biopsy (EmBx), and to investigate the association between myocardial fibrosis and clinical as well as functional markers in children after HTx. METHODS EmBx and CMR were performed on the same day. All specimens were stained with picrosirius red. The collagen volume fraction (CVF) was calculated as ratio of stained collagen area to total myocardial area on EmBx. Native T1 values and ECV were measured by CMR on a mid-ventricular short axis slice, using a modified look-locker inversion recovery approach. RESULTS Twenty patients (9.9 ± 6.2 years of age; 9 girls) after HTx were prospectively enrolled, at a median of 1.3 years (0.02-12.6 years) post HTx, and compared to 24 controls (13.9 ± 2.6 years of age; 12 girls). The mean histological CVF was 10.0 ± 3.4%. Septal native T1 times and ECV were higher in HTx patients compared to controls (1008 ± 32 ms vs 979 ± 24 ms, p < 0.005 and 0.30 ± 0.03 vs 0.22 ± 0.03, p < 0.0001, respectively). CVF showed a moderate correlation with native T1 (r = 0.53, p < 0.05) as well as ECV (r = 0.46, p < 0.05). Native T1 time, but not ECV and CVF, correlated with ischemia time (r = 0.46, p < 0.05). CONCLUSIONS CMR-derived fibrosis markers correlate with histological degree of fibrosis on EmBx in children after HTx. Further, native T1 times are associated with longer ischemia times.
Collapse
Affiliation(s)
- Seiko Ide
- Division of Cardiology, Department of Paediatrics, Labatt Family Heart Centre, The Hospital for Sick Children, University of Toronto, 555 University Avenue, Toronto, ON M5G 1X8 Canada
| | - Eugenie Riesenkampff
- Division of Cardiology, Department of Paediatrics, Labatt Family Heart Centre, The Hospital for Sick Children, University of Toronto, 555 University Avenue, Toronto, ON M5G 1X8 Canada
| | - David A. Chiasson
- Division of Pathology, Department of Paediatric Laboratory Medicine, The Hospital for Sick Children, University of Toronto, Toronto, ON Canada
| | - Anne I. Dipchand
- Division of Cardiology, Department of Paediatrics, Labatt Family Heart Centre, The Hospital for Sick Children, University of Toronto, 555 University Avenue, Toronto, ON M5G 1X8 Canada
| | - Paul F. Kantor
- Division of Cardiology, Department of Paediatrics, Labatt Family Heart Centre, The Hospital for Sick Children, University of Toronto, 555 University Avenue, Toronto, ON M5G 1X8 Canada
- Division of Cardiology, Department of Paediatrics, Stollery Children’s Hospital, Edmonton, AB Canada
| | - Rajiv R. Chaturvedi
- Division of Cardiology, Department of Paediatrics, Labatt Family Heart Centre, The Hospital for Sick Children, University of Toronto, 555 University Avenue, Toronto, ON M5G 1X8 Canada
| | - Shi-Joon Yoo
- Division of Cardiology, Department of Paediatrics, Labatt Family Heart Centre, The Hospital for Sick Children, University of Toronto, 555 University Avenue, Toronto, ON M5G 1X8 Canada
- Department of Diagnostic Imaging, The Hospital for Sick Children, University of Toronto, Toronto, ON Canada
| | - Lars Grosse-Wortmann
- Division of Cardiology, Department of Paediatrics, Labatt Family Heart Centre, The Hospital for Sick Children, University of Toronto, 555 University Avenue, Toronto, ON M5G 1X8 Canada
- Department of Diagnostic Imaging, The Hospital for Sick Children, University of Toronto, Toronto, ON Canada
| |
Collapse
|